1
|
Dai Q, Ain Q, Seth N, Rooney M, Zipprich A. Liver sinusoidal endothelial cells: Friend or foe in metabolic dysfunction- associated steatotic liver disease/metabolic dysfunction-associated steatohepatitis. Dig Liver Dis 2025; 57:493-503. [PMID: 39904692 DOI: 10.1016/j.dld.2025.01.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/27/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the predominant liver disease and is becoming the paramount contributor to end-stage liver disease and liver-related deaths. Liver sinusoidal endothelial cells (LSECs) located between the hepatic parenchyma and blood from viscera and gastrointestinal tract are the gatekeepers for the hepatic microenvironment and normal function. In normal physiological conditions, LSECs govern the substance exchange between hepatic parenchyma and blood through dynamic regulation of fenestration and maintain the quiescent state of Kupffer cells (KCs) and hepatic stellate cells. In MASLD, lipotoxicity, insulin resistance, gastrointestinal microbiota dysbiosis, and mechanical compression caused by fat-laden hepatocytes result in LSECs capillarization and dysfunction. The altered LSECs progressively shift from healer to injurer, exacerbating liver inflammation and advancing liver fibrosis. This review focuses on the deteriorative roles of LSECs and related molecular mechanisms involved in MASLD and their contribution to metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis development and progression. Furthermore, in this review, we propose that targeting LSECs dysfunction is a prospective therapeutic strategy to restore the physiological function of LSECs and mitigate MASLD progression.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Quratul Ain
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Navodita Seth
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Michael Rooney
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany
| | - Alexander Zipprich
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07747, Jena, Thuringia, Germany.
| |
Collapse
|
2
|
Jalal DI, Thurman JM, Smith RJ. Chronic kidney disease enhances alternative pathway activity: a new paradigm. J Clin Invest 2025; 135:e188353. [PMID: 40309771 PMCID: PMC12043098 DOI: 10.1172/jci188353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Reduced kidney function is associated with increased risk of cardiovascular disease in addition to kidney disease progression. Kidney disease is considered an inflammatory state, based on elevated levels of C-reactive protein and inflammatory cytokines. A key mediator of cardiovascular and kidney disease progression in the setting of reduced kidney function is systemic and vascular inflammation. However, the exact pathways that link chronic kidney disease (CKD) with inflammation remain incompletely understood. For decades it has been known that factor D, the main activator of the alternative complement pathway, is increased in the plasma of patients with reduced kidney function. Recent biomarker evidence suggests alternative pathway activation in this setting. CKD, therefore, seems to alter the balance of alternative pathway proteins, promoting inflammation and potentially exacerbating complement-mediated diseases and CKD-associated complications. In this manuscript, we review the impact of reduced kidney function on biomarkers of the alternative complement pathway and the implications of alternative pathway activation on cardiovascular disease and kidney disease progression. Importantly, we highlight the need for ongoing research efforts that may lead to opportunities to target the alternative pathway of complement withx the goal of improving kidney and cardiovascular outcomes in persons with reduced kidney function.
Collapse
Affiliation(s)
- Diana I. Jalal
- Division of Nephrology, Department of Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
- Center for Access and Delivery Research and Evaluation, Iowa City VA Health Care System, Iowa City, Iowa, USA
| | - Joshua M. Thurman
- Division of Renal Diseases and Hypertension, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Richard J.H. Smith
- Molecular Otolaryngology and Renal Research Laboratories, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
3
|
Gergues M, Bari R, Koppisetti S, Gosiewska A, Kang L, Hariri RJ. Senescence, NK cells, and cancer: navigating the crossroads of aging and disease. Front Immunol 2025; 16:1565278. [PMID: 40255394 PMCID: PMC12006071 DOI: 10.3389/fimmu.2025.1565278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Cellular senescence, a state of stable cell cycle arrest, acts as a double-edged sword in cancer biology. In young organisms, it acts as a barrier against tumorigenesis, but in the aging population, it may facilitate tumor growth and metastasis through the senescence-associated secretory phenotype (SASP). Natural killer (NK) cells play a critical role in the immune system, particularly in the surveillance, targeting, and elimination of malignant and senescent cells. However, age-related immunosenescence is characterized by declining NK cell function resulting in diminished ability to fight infection, eliminate senescent cells and suppress tumor development. This implies that preserving or augmenting NK cell function may be central to defense against age-related degenerative and malignant diseases. This review explores the underlying mechanisms behind these interactions, focusing on how aging influences the battle between the immune system and cancer, the implications of senescent NK cells in disease progression, and the potential of adoptive NK cell therapy as a countermeasure to these age-related immunological challenges.
Collapse
Affiliation(s)
| | | | | | | | - Lin Kang
- Research and Development, Celularity Inc., Florham Park, NJ, United States
| | | |
Collapse
|
4
|
Babcock MC, DuBose LE, Hildreth KL, Stauffer BL, Kohrt WM, Wenner MM, Moreau KL. Endothelial dysfunction in middle-aged and older men with low testosterone is associated with elevated circulating endothelin-1. Am J Physiol Regul Integr Comp Physiol 2025; 328:R253-R261. [PMID: 39887085 PMCID: PMC12121689 DOI: 10.1152/ajpregu.00218.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/07/2024] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
Low testosterone in middle-aged/older men contributes to accelerated vascular aging, including endothelial dysfunction. However, the mechanisms by which low testosterone affects endothelial dysfunction are not well understood. We sought to determine whether higher endothelin-1 (ET-1) levels are associated with reduced brachial artery flow-mediated dilation (FMD) in middle-aged/older men with low testosterone. Plasma ET-1 was quantified in 60 men categorized as young (n = 20, age = 30 ± 4 yr, testosterone = 510 ± 63 ng/dL), middle-aged/older with normal testosterone (n = 20, age = 59 ± 6 yr, testosterone = 512 ± 115 ng/dL), or middle-aged/older with low testosterone (n = 20, age = 60 ± 8 yr, testosterone = 265 ± 47 ng/dL). Endothelial function was determined via brachial artery FMD. Venous and arterial endothelial cells were harvested via endovascular biopsy in a subset of participants and stained for ET-1 expression. Middle-aged/older men with normal testosterone exhibited lower brachial artery FMD (5.7 ± 2.2%) compared with young men (7.3 ± 1.3%, P = 0.020), which was exaggerated in middle-aged/older men with low testosterone (4.0 ± 1.8%, P = 0.010 vs. middle-aged/older men with normal testosterone). Plasma ET-1 was not different between young (5.6 ± 0.9 ng/dL) and middle-aged/older men with normal testosterone (6.0 ± 1.4 ng/dL, P = 0.681) but was higher in middle-aged/older men with low testosterone (7.7 ± 2.8 ng/dL) compared with both groups (P < 0.001 vs. young men; P = 0.013 vs. middle-aged/older men with normal testosterone). There was no difference in venous (P = 0.616) or arterial (P = 0.222) endothelial cell ET-1 expression between groups. There was a significant inverse association between plasma ET-1 and FMD (r =-0.371, P = 0.004). These data suggest that the accelerated age-associated reduction in endothelial dysfunction in middle-aged/older men with low testosterone is related to higher circulating ET-1.NEW & NOTEWORTHY Middle-aged/older men with low testosterone have reduced vascular endothelial function compared with young and age-matched men with normal testosterone. In this manuscript, we demonstrate that men with low testosterone have higher plasma endothelin-1, which is associated with worse brachial artery flow-mediated dilation. The source of higher plasma endothelin-1 remains unknown; however, higher circulating endothelin-1 appears to be a mechanism contributing to reduced vascular endothelial function in men with low testosterone.
Collapse
Affiliation(s)
- Matthew C Babcock
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Lyndsey E DuBose
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kerry L Hildreth
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Brian L Stauffer
- Division of Cardiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Cardiology, Denver Health Medical Center, Denver, Colorado, United States
| | - Wendy M Kohrt
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Eastern Colorado, Denver, Colorado, United States
| | - Megan M Wenner
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| | - Kerrie L Moreau
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Eastern Colorado, Denver, Colorado, United States
| |
Collapse
|
5
|
Fekete M, Lehoczki A, Szappanos Á, Toth A, Mahdi M, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Ungvari Z. Cerebromicrovascular mechanisms contributing to long COVID: implications for neurocognitive health. GeroScience 2025; 47:745-779. [PMID: 39777702 PMCID: PMC11872997 DOI: 10.1007/s11357-024-01487-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Long COVID (also known as post-acute sequelae of SARS-CoV-2 infection [PASC] or post-COVID syndrome) is characterized by persistent symptoms that extend beyond the acute phase of SARS-CoV-2 infection, affecting approximately 10% to over 30% of those infected. It presents a significant clinical challenge, notably due to pronounced neurocognitive symptoms such as brain fog. The mechanisms underlying these effects are multifactorial, with mounting evidence pointing to a central role of cerebromicrovascular dysfunction. This review investigates key pathophysiological mechanisms contributing to cerebrovascular dysfunction in long COVID and their impacts on brain health. We discuss how endothelial tropism of SARS-CoV-2 and direct vascular infection trigger endothelial dysfunction, impaired neurovascular coupling, and blood-brain barrier disruption, resulting in compromised cerebral perfusion. Furthermore, the infection appears to induce mitochondrial dysfunction, enhancing oxidative stress and inflammation within cerebral endothelial cells. Autoantibody formation following infection also potentially exacerbates neurovascular injury, contributing to chronic vascular inflammation and ongoing blood-brain barrier compromise. These factors collectively contribute to the emergence of white matter hyperintensities, promote amyloid pathology, and may accelerate neurodegenerative processes, including Alzheimer's disease. This review also emphasizes the critical role of advanced imaging techniques in assessing cerebromicrovascular health and the need for targeted interventions to address these cerebrovascular complications. A deeper understanding of the cerebrovascular mechanisms of long COVID is essential to advance targeted treatments and mitigate its long-term neurocognitive consequences.
Collapse
Affiliation(s)
- Monika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary.
| | - Ágnes Szappanos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Mohamed Mahdi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, 4032, Debrecen, Hungary
- Infectology Clinic, University of Debrecen Clinical Centre, 4031, Debrecen, Hungary
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
6
|
Russo L, Babboni S, Andreassi MG, Daher J, Canale P, Del Turco S, Basta G. Treating Metabolic Dysregulation and Senescence by Caloric Restriction: Killing Two Birds with One Stone? Antioxidants (Basel) 2025; 14:99. [PMID: 39857433 PMCID: PMC11763027 DOI: 10.3390/antiox14010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Cellular senescence is a state of permanent cell cycle arrest accompanied by metabolic activity and characteristic phenotypic changes. This process is crucial for developing age-related diseases, where excessive calorie intake accelerates metabolic dysfunction and aging. Overnutrition disturbs key metabolic pathways, including insulin/insulin-like growth factor signaling (IIS), the mammalian target of rapamycin (mTOR), and AMP-activated protein kinase. The dysregulation of these pathways contributes to insulin resistance, impaired autophagy, exacerbated oxidative stress, and mitochondrial dysfunction, further enhancing cellular senescence and systemic metabolic derangements. On the other hand, dysfunctional endothelial cells and adipocytes contribute to systemic inflammation, reduced nitric oxide production, and altered lipid metabolism. Numerous factors, including extracellular vesicles, mediate pathological communication between the vascular system and adipose tissue, amplifying metabolic imbalances. Meanwhile, caloric restriction (CR) emerges as a potent intervention to counteract overnutrition effects, improve mitochondrial function, reduce oxidative stress, and restore metabolic balance. CR modulates pathways such as IIS, mTOR, and sirtuins, enhancing glucose and lipid metabolism, reducing inflammation, and promoting autophagy. CR can extend the health span and mitigate age-related diseases by delaying cellular senescence and improving healthy endothelial-adipocyte interactions. This review highlights the crosstalk between endothelial cells and adipocytes, emphasizing CR potential in counteracting overnutrition-induced senescence and restoring vascular homeostasis.
Collapse
Affiliation(s)
- Lara Russo
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Babboni
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Maria Grazia Andreassi
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El-Koura 100, Lebanon;
| | - Paola Canale
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Del Turco
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Giuseppina Basta
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| |
Collapse
|
7
|
Zhang J, Xia X, He S. Deciphering the causal association and underlying transcriptional mechanisms between telomere length and abdominal aortic aneurysm. Front Immunol 2024; 15:1438838. [PMID: 39234237 PMCID: PMC11371612 DOI: 10.3389/fimmu.2024.1438838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024] Open
Abstract
Background The purpose of this study is to investigate the causal effect and potential mechanisms between telomere length and abdominal aortic aneurysm (AAA). Methods Summary statistics of telomere length and AAA were derived from IEU open genome-wide association studies and FinnGen R9, respectively. Bi-directional Mendelian randomization (MR) analysis was conducted to reveal the causal relationship between AAA and telomere length. Three transcriptome datasets were retrieved from the Gene Expression Omnibus database and telomere related genes was down-loaded from TelNet. The overlapping genes of AAA related differentially expressed genes (DEGs), module genes, and telomere related genes were used for further investigation. Telomere related diagnostic biomarkers of AAA were selected with machine learning algorisms and validated in datasets and murine AAA model. The correlation between biomarkers and immune infiltration landscape was established. Results Telomere length was found to have a suggestive negative associations with AAA [IVW, OR 95%CI = 0.558 (0.317-0.701), P < 0.0001], while AAA showed no suggestive effect on telomere length [IVW, OR 95%CI = 0.997 (0.990-1.004), P = 0.4061]. A total of 40 genes was considered as telomere related DEGs of AAA. PLCH2, PRKCQ, and SMG1 were selected as biomarkers after multiple algorithms and validation. Immune infiltration analysis and single cell mRNA analysis revealed that PLCH2 and PRKCQ were mainly expressed on T cells, while SMG1 predominantly expressed on T cells, B cells, and monocytes. Murine AAA model experiments further validated the elevated expression of biomarkers. Conclusion We found a suggestive effect of telomere length on AAA and revealed the potential biomarkers and immune mechanism of telomere length on AAA. This may shed new light for diagnosis and therapeutics on AAA.
Collapse
Affiliation(s)
- Jiyu Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujie He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Mone P, Agyapong ED, Morciano G, Jankauskas SS, De Luca A, Varzideh F, Pinton P, Santulli G. Dysfunctional mitochondria elicit bioenergetic decline in the aged heart. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:13. [PMID: 39015481 PMCID: PMC11250775 DOI: 10.20517/jca.2023.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Aging represents a complex biological progression affecting the entire body, marked by a gradual decline in tissue function, rendering organs more susceptible to stress and diseases. The human heart holds significant importance in this context, as its aging process poses life-threatening risks. It entails macroscopic morphological shifts and biochemical changes that collectively contribute to diminished cardiac function. Among the numerous pivotal factors in aging, mitochondria play a critical role, intersecting with various molecular pathways and housing several aging-related agents. In this comprehensive review, we provide an updated overview of the functional role of mitochondria in cardiac aging.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
| | - Esther Densu Agyapong
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Stanislovas S. Jankauskas
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Vanvitelli University, Naples 80100, Italy
| | - Fahimeh Varzideh
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
- Department of Advanced Biomedical Sciences, “Federico II” University, International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples 80131, Italy
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
9
|
Liu S, He Y, Zhang Y, Zhang Z, Huang K, Deng L, Liao B, Zhong Y, Feng J. Targeting gut microbiota in aging-related cardiovascular dysfunction: focus on the mechanisms. Gut Microbes 2023; 15:2290331. [PMID: 38073096 PMCID: PMC10730151 DOI: 10.1080/19490976.2023.2290331] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
The global population is aging and age-related cardiovascular disease is increasing. Even after controlling for cardiovascular risk factors, readmission and mortality rates remain high. In recent years, more and more in-depth studies have found that the composition of the gut microbiota and its metabolites, such as trimethylamine N-oxide (TMAO), bile acids (BAs), and short-chain fatty acids (SCFAs), affect the occurrence and development of age-related cardiovascular diseases through a variety of molecular pathways, providing a new target for therapy. In this review, we discuss the relationship between the gut microbiota and age-related cardiovascular diseases, and propose that the gut microbiota could be a new therapeutic target for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Siqi Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yali Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Zhaolun Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Keming Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
10
|
Anastasiou M, Oikonomou E, Theofilis P, Papamikroulis GA, Gazouli M, Kalogeras K, Lygkoni S, Pesiridis T, Goliopoulou A, Papatheodoridi A, Psyrri A, Zagouri F, Siasos G, Tousoulis D. Prolonged impact of anti-cancer therapy on endothelial function and arterial stiffness in breast cancer patients. Vascul Pharmacol 2023; 152:107195. [PMID: 37455009 DOI: 10.1016/j.vph.2023.107195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Cardiotoxicity restricts anthracycline and trastuzumab treatment of Human Epidermal Growth Factor Receptor 2 positive early breast cancer. Endothelial dysfunction and arteriosclerosis are significant cardiovascular risk factors. OBJECTIVES We studied the effect of anthracycline-based chemotherapy, with or without trastuzumab, on endothelium and arteriosclerosis in patients with breast cancer. METHODS In this case-control study, 52 women with breast cancer and 104 women without breast cancer were examined longitudinally up to 15 months following (in the breast cancer group) initiation of chemotherapy. Arterial stiffness was evaluated through pulse wave velocity (PWV), while endothelial function via flow-mediated dilatation (FMD) at baseline (T0), 3 (T1), 6 (T2), and 15 (T3) months later. RESULTS There was no difference between subjects with breast cancer and control in PWV and FMD at baseline. Longitudinally, participants with breast cancer exhibited considerable impairment of PWV and FMD compared to the control group (p for interaction <0.001 for both parameters). In breast cancer patients, there was a significant increase from T0 to T3 in PWV (7.43 ± 1.68 m/s vs. 8.18 ± 2.00 m/s, p = 0.01) and decrease in FMD (6.95 ± 2.86% vs. 5.03 ± 2.83%, p = 0.006). The addition of trastuzumab in the treatment did not have any effect on PWV (p = 0.74) or FMD (p = 0.91). CONCLUSIONS In patients with breast cancer, there is progression of endothelial dysfunction and arteriosclerosis up to 15 months following initiation of anthracycline-based chemotherapy. Trastuzumab has no additive effect on endothelial function or arterial stiffness.
Collapse
Affiliation(s)
- Maria Anastasiou
- Section of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| | - Panagiotis Theofilis
- 1st Cardiology Department, "Hippokration" General Hospital, Medical School, University of Athens, 11527 Athens, Greece
| | - George Angelos Papamikroulis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Stavroula Lygkoni
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Theodoros Pesiridis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Alkistis Papatheodoridi
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, 11528 Athens, Greece
| | - Amanda Psyrri
- Section of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, 11528 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dimitris Tousoulis
- 1st Cardiology Department, "Hippokration" General Hospital, Medical School, University of Athens, 11527 Athens, Greece
| |
Collapse
|
11
|
Mengozzi A, de Ciuceis C, Dell'oro R, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Anyfanti P, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Mavraganis G, Montezano AC, Rios FJ, Winklewski PJ, Wolf J, Costantino S, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Triantafyllou A, Virdis A. The importance of microvascular inflammation in ageing and age-related diseases: a position paper from the ESH working group on small arteries, section of microvascular inflammation. J Hypertens 2023; 41:1521-1543. [PMID: 37382158 DOI: 10.1097/hjh.0000000000003503] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Microcirculation is pervasive and orchestrates a profound regulatory cross-talk with the surrounding tissue and organs. Similarly, it is one of the earliest biological systems targeted by environmental stressors and consequently involved in the development and progression of ageing and age-related disease. Microvascular dysfunction, if not targeted, leads to a steady derangement of the phenotype, which cumulates comorbidities and eventually results in a nonrescuable, very high-cardiovascular risk. Along the broad spectrum of pathologies, both shared and distinct molecular pathways and pathophysiological alteration are involved in the disruption of microvascular homeostasis, all pointing to microvascular inflammation as the putative primary culprit. This position paper explores the presence and the detrimental contribution of microvascular inflammation across the whole spectrum of chronic age-related diseases, which characterise the 21st-century healthcare landscape. The manuscript aims to strongly affirm the centrality of microvascular inflammation by recapitulating the current evidence and providing a clear synoptic view of the whole cardiometabolic derangement. Indeed, there is an urgent need for further mechanistic exploration to identify clear, very early or disease-specific molecular targets to provide an effective therapeutic strategy against the otherwise unstoppable rising prevalence of age-related diseases.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Raffaella Dell'oro
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - George Pavlidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Panagiota Anyfanti
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine
- Center of Translational Medicine
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University, Gdansk, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Cardiovascular Science, University College London, London, UK
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Francesco J Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | | | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Ignatios Ikonomidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | | | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
- Division of Medicine, Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site
- Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Areti Triantafyllou
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
12
|
Harrington JS, Ryter SW, Plataki M, Price DR, Choi AMK. Mitochondria in health, disease, and aging. Physiol Rev 2023; 103:2349-2422. [PMID: 37021870 PMCID: PMC10393386 DOI: 10.1152/physrev.00058.2021] [Citation(s) in RCA: 249] [Impact Index Per Article: 124.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are well known as organelles responsible for the maintenance of cellular bioenergetics through the production of ATP. Although oxidative phosphorylation may be their most important function, mitochondria are also integral for the synthesis of metabolic precursors, calcium regulation, the production of reactive oxygen species, immune signaling, and apoptosis. Considering the breadth of their responsibilities, mitochondria are fundamental for cellular metabolism and homeostasis. Appreciating this significance, translational medicine has begun to investigate how mitochondrial dysfunction can represent a harbinger of disease. In this review, we provide a detailed overview of mitochondrial metabolism, cellular bioenergetics, mitochondrial dynamics, autophagy, mitochondrial damage-associated molecular patterns, mitochondria-mediated cell death pathways, and how mitochondrial dysfunction at any of these levels is associated with disease pathogenesis. Mitochondria-dependent pathways may thereby represent an attractive therapeutic target for ameliorating human disease.
Collapse
Affiliation(s)
- John S Harrington
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | | | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - David R Price
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
13
|
Xue J, Zhang Z, Sun Y, Jin D, Guo L, Li X, Zhao D, Feng X, Qi W, Zhu H. Research Progress and Molecular Mechanisms of Endothelial Cells Inflammation in Vascular-Related Diseases. J Inflamm Res 2023; 16:3593-3617. [PMID: 37641702 PMCID: PMC10460614 DOI: 10.2147/jir.s418166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Endothelial cells (ECs) are widely distributed inside the vascular network, forming a vital barrier between the bloodstream and the walls of blood vessels. These versatile cells serve myriad functions, including the regulation of vascular tension and the management of hemostasis and thrombosis. Inflammation constitutes a cascade of biological responses incited by biological, chemical, or physical stimuli. While inflammation is inherently a protective mechanism, dysregulated inflammation can precipitate a host of vascular pathologies. ECs play a critical role in the genesis and progression of vascular inflammation, which has been implicated in the etiology of numerous vascular disorders, such as atherosclerosis, cardiovascular diseases, respiratory diseases, diabetes mellitus, and sepsis. Upon activation, ECs secrete potent inflammatory mediators that elicit both innate and adaptive immune reactions, culminating in inflammation. To date, no comprehensive and nuanced account of the research progress concerning ECs and inflammation in vascular-related maladies exists. Consequently, this review endeavors to synthesize the contributions of ECs to inflammatory processes, delineate the molecular signaling pathways involved in regulation, and categorize and consolidate the various models and treatment strategies for vascular-related diseases. It is our aspiration that this review furnishes cogent experimental evidence supporting the established link between endothelial inflammation and vascular-related pathologies, offers a theoretical foundation for clinical investigations, and imparts valuable insights for the development of therapeutic agents targeting these diseases.
Collapse
Affiliation(s)
- Jiaojiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yuting Sun
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Di Jin
- Department of Nephrology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Liming Guo
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiaochun Feng
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Haoyu Zhu
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| |
Collapse
|
14
|
Seegren PV, Harper LR, Downs TK, Zhao XY, Viswanathan SB, Stremska ME, Olson RJ, Kennedy J, Ewald SE, Kumar P, Desai BN. Reduced mitochondrial calcium uptake in macrophages is a major driver of inflammaging. NATURE AGING 2023:10.1038/s43587-023-00436-8. [PMID: 37277641 DOI: 10.1038/s43587-023-00436-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/09/2023] [Indexed: 06/07/2023]
Abstract
Mitochondrial dysfunction is linked to age-associated inflammation or inflammaging, but underlying mechanisms are not understood. Analyses of 700 human blood transcriptomes revealed clear signs of age-associated low-grade inflammation. Among changes in mitochondrial components, we found that the expression of mitochondrial calcium uniporter (MCU) and its regulatory subunit MICU1, genes central to mitochondrial Ca2+ (mCa2+) signaling, correlated inversely with age. Indeed, mCa2+ uptake capacity of mouse macrophages decreased significantly with age. We show that in both human and mouse macrophages, reduced mCa2+ uptake amplifies cytosolic Ca2+ oscillations and potentiates downstream nuclear factor kappa B activation, which is central to inflammation. Our findings pinpoint the mitochondrial calcium uniporter complex as a keystone molecular apparatus that links age-related changes in mitochondrial physiology to systemic macrophage-mediated age-associated inflammation. The findings raise the exciting possibility that restoring mCa2+ uptake capacity in tissue-resident macrophages may decrease inflammaging of specific organs and alleviate age-associated conditions such as neurodegenerative and cardiometabolic diseases.
Collapse
Affiliation(s)
- Philip V Seegren
- Pharmacology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Logan R Harper
- Pharmacology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Taylor K Downs
- Pharmacology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Xiao-Yu Zhao
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Microbiology, Immunology, and Cancer Biology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | - Marta E Stremska
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Microbiology, Immunology, and Cancer Biology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Rachel J Olson
- Pharmacology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Joel Kennedy
- Pharmacology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sarah E Ewald
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Microbiology, Immunology, and Cancer Biology Department, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Pankaj Kumar
- Biochemistry and Molecular Genetics Department, University of Virginia School of Medicine, Charlottesville, VA, USA
- University of Virginia, Bioinformatics Core, Charlottesville, VA, USA
| | - Bimal N Desai
- Pharmacology Department, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
15
|
Zhao W, Wang L, Wang Y, Yuan H, Zhao M, Lian H, Ma S, Xu K, Li Z, Yu G. Injured Endothelial Cell: A Risk Factor for Pulmonary Fibrosis. Int J Mol Sci 2023; 24:ijms24108749. [PMID: 37240093 DOI: 10.3390/ijms24108749] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
The pathological features of pulmonary fibrosis (PF) are the abnormal activation and proliferation of myofibroblasts and the extraordinary deposition of the extracellular matrix (ECM). However, the pathogenesis of PF is still indistinct. In recent years, many researchers have realized that endothelial cells had a crucial role in the development of PF. Studies have demonstrated that about 16% of the fibroblasts in the lung tissue of fibrotic mice were derived from endothelial cells. Endothelial cells transdifferentiated into mesenchymal cells via the endothelial-mesenchymal transition (E(nd)MT), leading to the excessive proliferation of endothelial-derived mesenchymal cells and the accumulation of fibroblasts and ECM. This suggested that endothelial cells, a significant component of the vascular barrier, played an essential role in PF. Herein, this review discusses E(nd)MT and its contribution to the activation of other cells in PF, which could provide new ideas for further understanding the source and activation mechanism of fibroblasts and the pathogenesis of PF.
Collapse
Affiliation(s)
- Weiming Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Yaxuan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Hongmei Yuan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Mengxia Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Hui Lian
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Shuaichen Ma
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Kai Xu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Zhongzheng Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Institute of Biomedical Science, College of Life Science, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
16
|
Li M, Wang D, Liu Z, Huang Y, Zhang Q, Pan C, Lin Y, Sun L, Zheng Y. Assessing the effects of aging on the renal endothelial cell landscape using single-cell RNA sequencing. Front Genet 2023; 14:1175716. [PMID: 37214419 PMCID: PMC10196692 DOI: 10.3389/fgene.2023.1175716] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/13/2023] [Indexed: 05/24/2023] Open
Abstract
Endothelial cells (ECs) with senescence-associated secretory phenotypes (SASP) have been identified as a key mechanism of aging that contributes to various age-related kidney diseases. In this study, we used single-cell RNA sequencing (scRNA-seq) to create a transcriptome atlas of murine renal ECs and identify transcriptomic changes that occur during aging. We identified seven different subtypes of renal ECs, with glomerular ECs and angiogenic ECs being the most affected by senescence. We confirmed our scRNA-seq findings by using double immunostaining for an EC marker (CD31) and markers of specialized EC phenotypes. Our analysis of the dynamics of capillary lineage development revealed a chronic state of inflammation and compromised glomerular function as prominent aging features. Additionally, we observed an elevated pro-inflammatory and pro-coagulant microenvironment in aged glomerular ECs, which may contribute to age-related glomerulosclerosis and renal fibrosis. Through intercellular communication analysis, we also identified changes in signaling involved in immune regulation that may contribute to a hostile microenvironment for renal homeostasis and function. Overall, our findings provide new insights into the mechanisms of aging in the renal endothelium and may pave the way for the discovery of diagnostic biomarkers and therapeutic interventions against age-related kidney diseases.
Collapse
Affiliation(s)
- Mengke Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Dongliang Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qikai Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Caineng Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Li Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yingfeng Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Carlini NA, Romanowski S, Rabalais EN, Kistler BM, Campbell MS, Krishnakumar IM, Harber MP, Fleenor BS. Coconut sugar derived from coconut inflorescence sap lowers systolic blood pressure and arterial stiffness in middle-aged and older adults: a pilot study. J Appl Physiol (1985) 2023; 134:508-514. [PMID: 36656985 DOI: 10.1152/japplphysiol.00394.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Nutraceutical-based interventions hold promise to reduce blood pressure (BP) and arterial stiffness, which are two cardiovascular disease (CVD) risk factors. However, the effects of coconut sap powder (CSP), an Asian sweetener and novel nutraceutical, on BP and arterial stiffness in middle-aged and older adults (MA/O, ≥45 yr) has yet to be established. We hypothesized CSP will decrease BP and arterial stiffness in MA/O adults. In a double-blind, randomized, placebo-controlled study design, 19 (age 55.3 ± 2.1 yr) MA/O adults completed measures of brachial and carotid BP, and arterial stiffness [carotid-femoral pulse wave velocity (cfPWV), common carotid artery (CCA) β-stiffness, compliance, distensibility, and Young's and Peterson's Elastic moduli] before and after 8 wk of CSP (1.5 g/day) or placebo (1.5 g/day). A two-way repeated-measures analysis of variance was used to compare group mean differences. Compared with placebo, CSP lowered brachial systolic BP (SBP) (CSP pre: 117.4 ± 2.9 vs. post: 109.0 ± 2.4 mmHg, P < 0.05), but not carotid SBP (P = 0.12). CSP also lowered Young's (CSP pre: 5,514.4 ± 1,115.4 vs. post: 3,690.6 ± 430.9 kPa) and Peterson's elastic moduli (CSP pre: 22.2 ± 4.4 vs. post: 19.2 ± 4.5 kPa) (P < 0.05, both). A trend for CSP to lower CCA β-stiffness (P = 0.06) and increase CCA compliance (P = 0.07) was also observed. Arterial stiffness assessed by cfPWV did not change (P > 0.05). No inflammatory or antioxidant biomarkers were affected by CSP. In summary, 8 wk of CSP lowers brachial SBP and CCA mechanical stiffness indicating a potential cardioprotective effect in MA/O adults.NEW & NOTEWORTHY Blood pressure (BP) and arterial stiffness are important predictors of cardiovascular health with aging. Nutraceuticals are an easy-to-implement lifestyle strategy demonstrating promise to effectively lower BP and arterial stiffness with aging and ultimately cardiovascular disease risk. We demonstrate that coconut sap powder (CSP), a traditional Asian sweetener, lowers brachial systolic BP and carotid artery mechanical stiffness in middle-aged and older (MA/O) adults. These findings provide initial evidence for the CSP-related cardioprotective effects in MA/O adults.
Collapse
Affiliation(s)
- Nicholas A Carlini
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Spencer Romanowski
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Emily N Rabalais
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Brandon M Kistler
- Department of Nutrition and Health Science, Ball State University, Muncie, Indiana, United States
| | - Marilyn S Campbell
- Department of Kinesiology and Health Promotion, University of Kentucky, Lexington, Kentucky, United States
| | | | - Matthew P Harber
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Bradley S Fleenor
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| |
Collapse
|
18
|
Pegadraju H, Abby Thomas J, Kumar R. Mechanistic and therapeutic role of Drp1 in the pathogenesis of stroke. Gene 2023; 855:147130. [PMID: 36543307 DOI: 10.1016/j.gene.2022.147130] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Stroke had emerged as one of the leading causes of death and long-term disability across the globe. Emerging evidence suggests a significant increase in the incidence of stroke with age, which is further expected to increase dramatically owing to an ever-expanding elderly population. The current situation imposes a significant burden on the healthcare system and requires a deeper understanding of the underlying mechanisms and development of novel interventions. It is well established that mitochondrial dysfunction plays a pivotal role in the onset of stroke. Dynamin-related protein 1 (Drp1), is a key regulator of mitochondria fission, and plays a crucial role during the pathogenesis of stroke. Drp1 protein levels significantly increase after stroke potentially in a p38 mitogen-activated protein kinases (MAPK) dependent manner. Protein phosphatase 2A (PP2A) facilitate mitochondrial fission and cell death by dephosphorylating the mitochondrial fission enzyme Drp1 at the inhibitory phosphorylation site serine 637. Outer mitochondrial membrane A-Kinase Anchoring Proteins 1 (AKAP 1) and protein kinase A complex (PKA) complex inhibits Drp1-dependent mitochondrial fission by phosphorylating serine 637. Drp1 activation promotes the release of cytochrome C from mitochondria and therefore leads to apoptosis. In addition, Drp1 activation inhibits mitochondrial glutathione dependent free radical scavenging, which further enhances the ROS level and exacerbate mitochondrial dysfunction. Drp1 translocate p53 to mitochondrial membrane and leads to mitochondria-related necrosis. The current review article discusses the possible mechanistic pathways by which Drp1 can influence the pathogenesis of stroke. Besides, it will describe various inhibitors for Drp1 and their potential role as therapeutics for stroke in the future.
Collapse
Affiliation(s)
- Himaja Pegadraju
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Joshua Abby Thomas
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Rahul Kumar
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India.
| |
Collapse
|
19
|
Murray KO, Mahoney SA, Venkatasubramanian R, Seals DR, Clayton ZS. Aging, aerobic exercise, and cardiovascular health: Barriers, alternative strategies and future directions. Exp Gerontol 2023; 173:112105. [PMID: 36731386 PMCID: PMC10068966 DOI: 10.1016/j.exger.2023.112105] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
Age-associated cardiovascular (CV) dysfunction, namely arterial dysfunction, is a key antecedent to the development of CV disease (CVD). Arterial dysfunction with aging is characterized by impaired vascular endothelial function and stiffening of the large elastic arteries, each of which is an independent predictor of CVD. These processes are largely mediated by an excess production of reactive oxygen species (ROS) and an increase in chronic, low-grade inflammation that ultimately leads to a reduction in bioavailability of the vasodilatory molecule nitric oxide. Additionally, there are other fundamental aging mechanisms that may contribute to excessive ROS and inflammation termed the "hallmarks of aging"; these additional mechanisms of arterial dysfunction may represent therapeutic targets for improving CV health with aging. Aerobic exercise is the most well-known and effective intervention to prevent and treat the effects of aging on CV dysfunction. However, the majority of mid-life and older (ML/O) adults do not meet recommended exercise guidelines due to traditional barriers to aerobic exercise, such as reduced leisure time, motivation, or access to fitness facilities. Therefore, it is a biomedical research priority to develop and implement time- and resource-efficient alternative strategies to aerobic exercise to reduce the burden of CVD in ML/O adults. Alternative strategies that mimic or are inspired by aerobic exercise, that target pathways specific to the fundamental mechanisms of aging, represent a promising approach to accomplish this goal.
Collapse
Affiliation(s)
- Kevin O Murray
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America
| | - Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America
| | | | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America.
| |
Collapse
|
20
|
Wang D, Li M, Ling J, Chen S, Zhang Q, Liu Z, Huang Y, Pan C, Lin Y, Shi Z, Zhang P, Zheng Y. Assessing the effects of aging on the liver endothelial cell landscape using single-cell RNA sequencing. Hepatol Commun 2023; 7:e0021. [PMID: 36724124 PMCID: PMC9894352 DOI: 10.1097/hc9.0000000000000021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/25/2022] [Indexed: 02/02/2023] Open
Abstract
Endothelial cell (EC) function declines with age and contributes to the development of many vascular-related disease processes. Currently, the effects of aging on the molecular regulatory mechanisms of liver ECs have not been fully elucidated. Here, we employed single-cell RNA sequencing to map the transcriptome of ECs and analyzed their relationship with aging. We identified 8 different EC subtypes, interestingly, 2 of which were specially expressed in aged mice ECs namely aged capillary ECs (Aged ECs) and pro-inflammation capillary ECs (Proinfla.ECs). Double immunostaining for an EC marker (Cd31) and a marker of these specialized EC phenotypes confirmed the single-cell RNA sequencing data. Gene ontology analysis revealed that Aged ECs and Proinfla.ECs were associated with inflammatory response. Then we found that liver proliferating capillary ECs (Prolife.ECs) were most affected by senescence. Single-cell transcript analysis suggests that Prolife.ECs and angiogenic capillary ECs may form a poor microenvironment that promotes angiogenesis and tumorigenesis. Pseudo-temporal trajectories revealed that Prolife.ECs have different differentiation pathways in young and aged mice. In aged mice, Prolife.ECs could specifically differentiate into an unstable state, which was mainly composed of angiogenic capillary ECs. Intercellular communication revealed inflammatory activation in old group. Overall, this work compared the single-cell RNA profiles of liver ECs in young and aged mice. These findings provide a new insight into liver aging and its molecular mechanisms, and further exploration of Aged ECs and Proinfla.ECs may help to elucidate the molecular mechanisms associated with senescence.
Collapse
|
21
|
Gao Q, Yang L, Teng F, Guo S. Peripheral blood monocyte status is a predictor for judging occurrence and development on sepsis in older adult population: a case control study. BMC Emerg Med 2023; 23:11. [PMID: 36721090 PMCID: PMC9890732 DOI: 10.1186/s12873-023-00779-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/19/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Peripheral blood monocytes are important immune modulatory cells that change during aging. Previous studies on sepsis and monocytes did not distinguish between age groups, especially in the older adult population. The mechanisms of monocyte subsets and function are not well-understood in the aging context with sepsis. METHODS Monocyte subsets were measured using flow cytometry in 80 sepsis patients and 40 healthy controls. Plasma cytokine levels were measured using cytokine antibody arrays. RESULTS The percentage of MO3 (CD14 + CD16 + +)/monocytes was higher in sepsis patients than in controls (P = 0.011), whereas the percentage of MO1 (CD14 + + CD16 -)/monocytes was higher in septic shock patients and 28-day death group than in those without shock and 28-day survival group (P = 0.034, 0.038). Logistic regression analysis showed that the percentage of MO3/monocytes (OR = 1.120, P = 0.046) and plasma level of monocyte chemoattractant protein (MCP)-1 (OR = 1.006, P = 0.023) were independently associated with the occurrence of sepsis, whereas the percentage of MO1/monocytes (OR = 1.255, P = 0.048) was independently associated with septic shock. The receiver operating characteristic (ROC) curve showed that the area under the curve (AUC) of MO3/monocyte percentage in combination with MCP-1 plasma level (AUC = 0.799) for predicting sepsis was higher than that of each parameter alone (P < 0.001). The AUC of MO1/monocyte percentage with the value 0.706 (P = 0.003) was lower than the AUC of SOFA (sequential organ failure assessment) score with the value 0.966 (P < 0.001) for predicting septic shock, but the value of the two AUCs were similar for predicting 28-day mortality (AUC = 0.705, 0.827; P = 0.020, P < 0.001). The AUC of MO1/monocytes percentage in combination with SOFA score for predicting 28-day mortality was higher than that of each parameter alone (AUC = 0.867, P < 0.001). Using a cut-off of 58.5% (for MO1/monocytes determined by ROC) could discriminate between survivors and non-survivors on Kaplan-Meier curves for 28-day mortality with a positive predictive value of 77.4%. CONCLUSION The MO3/monocyte percentage and plasma MCP-1 level were independent predictors of sepsis occurrence, whereas the percentage of MO1/monocytes was an independent predictor of prognosis in the Chinese Han older adult population. TRIAL REGISTRATION Registration number: ChiCTR2200061490, date of registration: 2022-6-26 (retrospectively registered).
Collapse
Affiliation(s)
- Qian Gao
- grid.414367.3Emergency Department, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Yangfangdian, Haidian District, Beijing, 100038 China
| | - Li Yang
- grid.414367.3Emergency Department, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Yangfangdian, Haidian District, Beijing, 100038 China
| | - Fei Teng
- grid.411607.5Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Road of Worker’s Stadium, Chaoyang District, Beijing, 100020 China
| | - Shu‑Bin Guo
- grid.411607.5Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Road of Worker’s Stadium, Chaoyang District, Beijing, 100020 China
| |
Collapse
|
22
|
Association between atherogenic risk-modulating proteins and endothelium-dependent flow-mediated dilation in coronary artery disease patients. Eur J Appl Physiol 2023; 123:367-380. [PMID: 36305972 PMCID: PMC9894982 DOI: 10.1007/s00421-022-05040-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 09/04/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Endothelial dysfunction is an early and integral event in the development of atherosclerosis and coronary artery disease (CAD). Reduced NO bioavailability, oxidative stress, vasoconstriction, inflammation and senescence are all implicated in endothelial dysfunction. However, there are limited data examining associations between these pathways and direct in vivo bioassay measures of endothelial function in CAD patients. This study aimed to examine the relationships between in vivo measures of vascular function and the expression of atherogenic risk-modulating proteins in endothelial cells (ECs) isolated from the radial artery of CAD patients. METHODS Fifty-six patients with established CAD underwent trans-radial catheterization. Prior to catheterization, radial artery vascular function was assessed using a) flow-mediated dilation (FMD), and b) exercise-induced dilation in response to handgrip (HE%). Freshly isolated ECs were obtained from the radial artery during catheterization and protein content of eNOS, NAD(P)H oxidase subunit NOX2, NFκB, ET-1 and the senescence markers p53, p21 and p16 were evaluated alongside nitrotyrosine abundance and eNOS Ser1177 phosphorylation. RESULTS FMD was positively associated with eNOS Ser1177 phosphorylation (r = 0.290, P = 0.037), and protein content of p21 (r = 0.307, P = 0.027) and p16 (r = 0.426, P = 0.002). No associations were found between FMD and markers of oxidative stress, vasoconstriction or inflammation. In contrast to FMD, HE% was not associated with any of the EC proteins. CONCLUSION These data revealed a difference in the regulation of endothelium-dependent vasodilation measured in vivo between patients with CAD compared to previously reported data in subjects without a clinical diagnosis, suggesting that eNOS Ser1177 phosphorylation may be the key to maintain vasodilation in CAD patients.
Collapse
|
23
|
Hamel KM, King CT, Cavalier MB, Liimatta KQ, Rozanski GL, King TA, Lam M, Bingham GC, Byrne CE, Xing D, Collins-Burow BM, Burow ME, Belgodere JA, Bratton MR, Bunnell BA, Martin EC. Breast Cancer-Stromal Interactions: Adipose-Derived Stromal/Stem Cell Age and Cancer Subtype Mediated Remodeling. Stem Cells Dev 2022; 31:604-620. [PMID: 35579936 PMCID: PMC9595652 DOI: 10.1089/scd.2021.0279] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/16/2022] [Indexed: 10/18/2022] Open
Abstract
Adipose tissue is characterized as an endocrine organ that acts as a source of hormones and paracrine factors. In diseases such as cancer, endocrine and paracrine signals from adipose tissue contribute to cancer progression. Young individuals with estrogen receptor-alpha positive (ER-α+) breast cancer (BC) have an increased resistance to endocrine therapies, suggesting that alternative estrogen signaling is activated within these cells. Despite this, the effects of stromal age on the endocrine response in BC are not well defined. To identify differences between young and aged ER-α+ breast tumors, RNA sequencing data were obtained from The Cancer Genome Atlas. Analysis revealed enrichment of matrix and paracrine factors in young (≤40 years old) patients compared to aged (≥65 years old) tumor samples. Adipose-derived stromal/stem cells (ASCs) from noncancerous lipoaspirate of young and aged donors were evaluated for alterations in matrix production and paracrine secreted factors to determine if the tumor stroma could alter estrogen signaling. Young and aged ASCs demonstrated comparable proliferation, differentiation, and matrix production, but exhibited differences in the expression levels of inflammatory cytokines (Interferon gamma, interleukin [IL]-8, IL-10, Tumor necrosis factor alpha, IL-2, and IL-6). Conditioned media (CM)-based experiments showed that young ASC donor age elevated endocrine response in ER-α+ BC cell lines. MCF-7 ER-α+ BC cell line treated with secreted factors from young ASCs had enhanced ER-α regulated genes (PGR and SDF-1) compared to MCF-7 cells treated with aged ASC CM. Western blot analysis demonstrated increased activation levels of p-ER ser-167 in the MCF-7 cell line treated with young ASC secreted factors. To determine if ER-α+ BC cells heightened the cytokine release in ASCs, ASCs were stimulated with MCF-7-derived CM. Results demonstrated no change in growth factors or cytokines when treated with the ER-α+ secretome. In contrast to ER-α+ CM, the ER-α negative MDA-MB-231 derived CM demonstrated increased stimulation of pro-inflammatory cytokines in ASCs. While there was no observed change in the release of selected paracrine factors, MCF-7 cells did induce matrix production and a pro-adipogenic lineage commitment. The adipogenesis was evident by increased collagen content through Sirius Red/Fast Green Collagen stain, lipid accumulation evident by Oil Red O stain, and significantly increased expression in PPARγ mRNA expression. The data from this study provide evidence suggesting more of a subtype-dependent than an age-dependent difference in stromal response to BC, suggesting that this signaling is not heightened by reciprocal signals from ER-α+ BC cell lines. These results are important in understanding the mechanisms of estrogen signaling and the dynamic and reciprocal nature of cancer cell-stromal cell crosstalk that can lead to tumor heterogeneity and variance in response to therapy.
Collapse
Affiliation(s)
- Katie M. Hamel
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Connor T. King
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Maryn B. Cavalier
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Kara Q. Liimatta
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Grace L. Rozanski
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Timothy A. King
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Meggie Lam
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Grace C. Bingham
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - C. Ethan Byrne
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Diensn Xing
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bridgette M. Collins-Burow
- Section of Hematology and Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Matthew E. Burow
- Section of Hematology and Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jorge A. Belgodere
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | | | - Bruce A. Bunnell
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Sciences Center, Fort Worth, Texas, USA
| | - Elizabeth C. Martin
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
24
|
Murray KO, Berryman-Maciel M, Darvish S, Coppock ME, You Z, Chonchol M, Seals DR, Rossman MJ. Mitochondrial-targeted antioxidant supplementation for improving age-related vascular dysfunction in humans: A study protocol. Front Physiol 2022; 13:980783. [PMID: 36187760 PMCID: PMC9520456 DOI: 10.3389/fphys.2022.980783] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Cardiovascular disease (CVD) is the leading cause of death worldwide and aging is the primary risk factor for the development of CVD. The increased risk of CVD with aging is largely mediated by the development of vascular dysfunction. Excessive production of mitochondrial reactive oxygen species (mtROS) is a key mechanism of age-related vascular dysfunction. Therefore, establishing the efficacy of therapies to reduce mtROS to improve vascular function with aging is of high biomedical importance. Previously, in a small, randomized, crossover-design pilot clinical trial, our laboratory obtained initial evidence that chronic oral supplementation with the mitochondrial-targeted antioxidant MitoQ improves vascular function in healthy older adults. Here, we describe the protocol for an ongoing R01-funded phase IIa clinical trial to establish the efficacy of MitoQ as a therapy to improve vascular function in older adults (ClinicalTrials.gov Identifier: NCT04851288).Outcomes: The primary outcome of the study is nitric oxide (NO)-mediated endothelium-dependent dilation (EDD) as assessed by brachial artery flow-mediated dilation (FMDBA). Secondary outcomes include mtROS-mediated suppression of EDD, aortic stiffness as measured by carotid-femoral pulse wave velocity, carotid compliance and β-stiffness index, and intima media thickness. Other outcomes include the assessment of endothelial mitochondrial health and oxidative stress in endothelial cells obtained by endovascular biopsy; the effect of altered circulating factors following MitoQ treatment on endothelial cell NO bioavailability and whole cell and mitochondrial reactive oxygen species production ex vivo; and circulating markers of oxidative stress, antioxidant status, and inflammation.Methods: We are conducting a randomized, placebo-controlled, double-blind, parallel group, phase IIa clinical trial in 90 (45/group) healthy older men and women 60 years of age or older. Participants complete baseline testing and are then randomized to either 3 months of oral MitoQ (20 mg; once daily) or placebo supplementation. Outcome measures are assessed at the midpoint of treatment, i.e., 6 weeks, and again at the conclusion of treatment.Discussion: This study is designed to establish the efficacy of chronic supplementation with the mitochondrial-targeted antioxidant MitoQ for improving vascular endothelial function and reducing large elastic artery stiffness in older adults, and to investigate the mechanisms by which MitoQ supplementation improves endothelial function.
Collapse
Affiliation(s)
- Kevin O. Murray
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Morgan Berryman-Maciel
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Sanna Darvish
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - McKinley E. Coppock
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Zhiying You
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Douglas R. Seals
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Matthew J. Rossman
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
- *Correspondence: Matthew J. Rossman,
| |
Collapse
|
25
|
Craighead DH, Freeberg KA, McCarty NP, Rossman MJ, Moreau KL, You Z, Chonchol M, Seals DR. Inspiratory muscle strength training for lowering blood pressure and improving endothelial function in postmenopausal women: comparison with “standard of care” aerobic exercise. Front Physiol 2022; 13:967478. [PMID: 36105300 PMCID: PMC9465043 DOI: 10.3389/fphys.2022.967478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background: High blood pressure (BP), particularly systolic BP (SBP), is the major modifiable risk factor for cardiovascular diseases and related disorders of aging. SBP increases markedly with aging in women such that the prevalence of above-normal SBP (i.e., ≥120 mmHg) in postmenopausal women exceeds rates in age-matched men. This increase in SBP is associated with vascular endothelial dysfunction, mediated by excessive reactive oxygen species-induced oxidative stress and consequent reductions in nitric oxide bioavailability. Moderate-intensity aerobic exercise is a recommended lifestyle strategy for reducing SBP. However, adherence to aerobic exercise guidelines among postmenopausal women is low (<30%) and aerobic exercise does not consistently enhance endothelial function in estrogen-deficient postmenopausal women. High-resistance inspiratory muscle strength training (IMST) is a time-efficient, adherable lifestyle intervention that involves inhaling against resistance through a handheld device (30 breaths/day). Here, we present the protocol for a randomized controlled trial investigating the efficacy of 3 months of high-resistance IMST compared to guideline-based, “standard-of-care” aerobic exercise training for decreasing SBP and improving endothelial function in estrogen-deficient postmenopausal women with above-normal SBP (120–159 mmHg) at baseline (ClinicalTrials.gov Identifier: NCT05000515). Methods: A randomized, single-blind, parallel-group design clinical trial will be conducted in 72 (36/group) estrogen-deficient postmenopausal women with above-normal SBP. Participants will complete baseline testing and then be randomized to either 3 months of high-resistance IMST (30 breaths/day, 6 days/week, 75% maximal inspiratory pressure) or moderate-intensity aerobic exercise training (brisk walking 25 min/day, 6 days/week, 40–60% heart rate reserve). Outcome measures will be assessed after 3 months of either intervention. Following end-intervention testing, participants will abstain from their assigned intervention for 6 weeks, after which BP and endothelial function will be assessed to evaluate the potential persistent effects of the intervention on the primary and secondary outcomes. Discussion: This study is designed to compare the effectiveness of time-efficient, high-resistance IMST to guideline-based aerobic exercise training for lowering SBP and improving endothelial function, and interrogating potential mechanisms of action, in estrogen-deficient postmenopausal women. Clinical Trial Registration:ClinicalTrials.gov, Identifier: NCT05000515.
Collapse
Affiliation(s)
- Daniel H. Craighead
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Kaitlin A. Freeberg
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Narissa P. McCarty
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Matthew J. Rossman
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Kerrie L. Moreau
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Veterans Affairs Eastern Colorado Geriatric Research, Educational and Clinical Center, Denver, CO, United States
| | - Zhiying You
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Douglas R. Seals
- Integrative Physiology of Aging Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
- *Correspondence: Douglas R. Seals,
| |
Collapse
|
26
|
Tyrrell DJ, Chen J, Li BY, Wood SC, Rosebury-Smith W, Remmer HA, Jiang L, Zhang M, Salmon M, Ailawadi G, Yang B, Goldstein DR. Aging Alters the Aortic Proteome in Health and Thoracic Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2022; 42:1060-1076. [PMID: 35510553 PMCID: PMC9339483 DOI: 10.1161/atvbaha.122.317643] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Aging enhances most chronic diseases but its impact on human aortic tissue in health and in thoracic aortic aneurysms (TAA) remains unclear. METHODS We employed a human aortic biorepository of healthy specimens (n=17) and those that underwent surgical repair for TAA (n=20). First, we performed proteomics comparing aortas of healthy donors to aneurysmal specimens, in young (ie, <60 years of age) and old (ie, ≥60 years of age) subjects. Second, we measured proteins, via immunoblotting, involved in mitophagy (ie, Parkin) and also mitochondrial-induced inflammatory pathways, specifically TLR (toll-like receptor) 9, STING (stimulator of interferon genes), and IFN (interferon)-β. RESULTS Proteomics revealed that aging transformed the aorta both quantitatively and qualitatively from health to TAA. Whereas young aortas exhibited an enrichment of immunologic processes, older aortas exhibited an enrichment of metabolic processes. Immunoblotting revealed that the expression of Parkin directly correlated to subject age in health but inversely to subject age in TAA. In TAA, but not in health, phosphorylation of STING and the expression of IFN-β was impacted by aging regardless of whether subjects had bicuspid or tricuspid valves. In subjects with bicuspid valves and TAAs, TLR9 expression positively correlated with subject age. Interestingly, whereas phosphorylation of STING was inversely correlated with subject age, IFN-β positively correlated with subject age. CONCLUSIONS Aging transforms the human aortic proteome from health to TAA, leading to a differential regulation of biological processes. Our results suggest that the development of therapies to mitigate vascular diseases including TAA may need to be modified depending on subject age.
Collapse
Affiliation(s)
| | - Judy Chen
- Department of Internal Medicine, University of Michigan, USA,Program on Immunology, University of Michigan, USA
| | - Benjamin Y. Li
- Department of Internal Medicine, University of Michigan, USA
| | - Sherri C. Wood
- Department of Internal Medicine, University of Michigan, USA
| | | | | | - Longtan Jiang
- Department of Cardiac Surgery, University of Michigan, USA
| | - Min Zhang
- Department of Biostatistics, University of Michigan, USA
| | - Morgan Salmon
- Department of Cardiac Surgery, University of Michigan, USA
| | - Gorav Ailawadi
- Department of Cardiac Surgery, University of Michigan, USA
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan, USA
| | - Daniel R. Goldstein
- Department of Internal Medicine, University of Michigan, USA,Program on Immunology, University of Michigan, USA,Department of Microbiology and Immunology, University of Michigan, USA
| |
Collapse
|
27
|
Kuczmarski AV, Welti LM, Moreau KL, Wenner MM. ET-1 as a Sex-Specific Mechanism Impacting Age-Related Changes in Vascular Function. FRONTIERS IN AGING 2022; 2:727416. [PMID: 35822003 PMCID: PMC9261354 DOI: 10.3389/fragi.2021.727416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/13/2021] [Indexed: 01/30/2023]
Abstract
Aging is a primary risk factor for cardiovascular disease (CVD), which is the leading cause of death in developed countries. Globally, the population of adults over the age of 60 is expected to double by the year 2050. CVD prevalence and mortality rates differ between men and women as they age in part due to sex-specific mechanisms impacting the biological processes of aging. Measures of vascular function offer key insights into cardiovascular health. Changes in vascular function precede changes in CVD prevalence rates in men and women and with aging. A key mechanism underlying these changes in vascular function is the endothelin (ET) system. Studies have demonstrated sex and sex hormone effects on endothelin-1 (ET-1), and its receptors ETA and ETB. However, with aging there is a dysregulation of this system resulting in an imbalance between vasodilation and vasoconstriction. Thus, ET-1 may play a role in the sex differences observed with vascular aging. While most research has been conducted in pre-clinical animal models, we describe more recent translational data in humans showing that the ET system is an important regulator of vascular dysfunction with aging and acts through sex-specific ET receptor mechanisms. In this review, we present translational evidence (cell, tissue, animal, and human) that the ET system is a key mechanism regulating sex-specific changes in vascular function with aging, along with therapeutic interventions to reduce ET-mediated vascular dysfunction associated with aging. More knowledge on the factors responsible for the sex differences with vascular aging allow for optimized therapeutic strategies to attenuate CVD risk in the expanding aging population.
Collapse
Affiliation(s)
- Andrew V Kuczmarski
- University of Delaware, Kinesiology and Applied Physiology, Newark, DE, United States
| | - Laura M Welti
- University of Delaware, Kinesiology and Applied Physiology, Newark, DE, United States
| | - Kerrie L Moreau
- University of Colorado, Anschutz Medical Campus, Aurora, CO, United States.,Denver Veterans Administrative Medical Center, Geriatric Research Education and Clinical Center, Aurora, CO, United States
| | - Megan M Wenner
- University of Delaware, Kinesiology and Applied Physiology, Newark, DE, United States
| |
Collapse
|
28
|
Lázničková P, Bendíčková K, Kepák T, Frič J. Immunosenescence in Childhood Cancer Survivors and in Elderly: A Comparison and Implication for Risk Stratification. FRONTIERS IN AGING 2022; 2:708788. [PMID: 35822014 PMCID: PMC9261368 DOI: 10.3389/fragi.2021.708788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022]
Abstract
The population of childhood cancer survivors (CCS) has grown rapidly in recent decades. Although cured of their original malignancy, these individuals are at increased risk of serious late effects, including age-associated complications. An impaired immune system has been linked to the emergence of these conditions in the elderly and CCS, likely due to senescent immune cell phenotypes accompanied by low-grade inflammation, which in the elderly is known as "inflammaging." Whether these observations in the elderly and CCS are underpinned by similar mechanisms is unclear. If so, existing knowledge on immunosenescent phenotypes and inflammaging might potentially serve to benefit CCS. We summarize recent findings on the immune changes in CCS and the elderly, and highlight the similarities and identify areas for future research. Improving our understanding of the underlying mechanisms and immunosenescent markers of accelerated immune aging might help us to identify individuals at increased risk of serious health complications.
Collapse
Affiliation(s)
- Petra Lázničková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Tomáš Kepák
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Pediatric Oncology, University Hospital Brno, Brno, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
29
|
Xu H, Li S, Liu YS. Nanoparticles in the diagnosis and treatment of vascular aging and related diseases. Signal Transduct Target Ther 2022; 7:231. [PMID: 35817770 PMCID: PMC9272665 DOI: 10.1038/s41392-022-01082-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 11/09/2022] Open
Abstract
Aging-induced alternations of vasculature structures, phenotypes, and functions are key in the occurrence and development of vascular aging-related diseases. Multiple molecular and cellular events, such as oxidative stress, mitochondrial dysfunction, vascular inflammation, cellular senescence, and epigenetic alterations are highly associated with vascular aging physiopathology. Advances in nanoparticles and nanotechnology, which can realize sensitive diagnostic modalities, efficient medical treatment, and better prognosis as well as less adverse effects on non-target tissues, provide an amazing window in the field of vascular aging and related diseases. Throughout this review, we presented current knowledge on classification of nanoparticles and the relationship between vascular aging and related diseases. Importantly, we comprehensively summarized the potential of nanoparticles-based diagnostic and therapeutic techniques in vascular aging and related diseases, including cardiovascular diseases, cerebrovascular diseases, as well as chronic kidney diseases, and discussed the advantages and limitations of their clinical applications.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.,Institute of Aging and Age-related Disease Research, Central South University, 410011, Changsha, Hunan, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.,Institute of Aging and Age-related Disease Research, Central South University, 410011, Changsha, Hunan, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China. .,Institute of Aging and Age-related Disease Research, Central South University, 410011, Changsha, Hunan, China.
| |
Collapse
|
30
|
Craighead DH, Freeberg KA, Maurer GS, Myers VH, Seals DR. Translational Potential of High-Resistance Inspiratory Muscle Strength Training. Exerc Sport Sci Rev 2022; 50:107-117. [PMID: 35394978 PMCID: PMC9203907 DOI: 10.1249/jes.0000000000000293] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Age-associated cardiovascular (CV) dysfunction increases the risk for CV diseases. Aerobic exercise training can improve CV function, but only a minority of adults meet aerobic exercise guidelines. High-resistance inspiratory muscle strength training is a time-efficient lifestyle intervention that may promote adherence and improve CV function. However, further investigation is needed to translate inspiratory muscle strength training into the public health domain.
Collapse
Affiliation(s)
- Daniel H. Craighead
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Kaitlin A. Freeberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Grace S. Maurer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Douglas R. Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
31
|
Pierce GL, Coutinho TA, DuBose LE, Donato AJ. Is It Good to Have a Stiff Aorta with Aging? Causes and Consequences. Physiology (Bethesda) 2022; 37:154-173. [PMID: 34779281 PMCID: PMC8977146 DOI: 10.1152/physiol.00035.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/09/2023] Open
Abstract
Aortic stiffness increases with advancing age, more than doubling during the human life span, and is a robust predictor of cardiovascular disease (CVD) clinical events independent of traditional risk factors. The aorta increases in diameter and length to accommodate growing body size and cardiac output in youth, but in middle and older age the aorta continues to remodel to a larger diameter, thinning the pool of permanent elastin fibers, increasing intramural wall stress and resulting in the transfer of load bearing onto stiffer collagen fibers. Whereas aortic stiffening in early middle age may be a compensatory mechanism to normalize intramural wall stress and therefore theoretically "good" early in the life span, the negative clinical consequences of accelerated aortic stiffening beyond middle age far outweigh any earlier physiological benefit. Indeed, aortic stiffness and the loss of the "windkessel effect" with advancing age result in elevated pulsatile pressure and flow in downstream microvasculature that is associated with subclinical damage to high-flow, low-resistance organs such as brain, kidney, retina, and heart. The mechanisms of aortic stiffness include alterations in extracellular matrix proteins (collagen deposition, elastin fragmentation), increased arterial tone (oxidative stress and inflammation-related reduced vasodilators and augmented vasoconstrictors; enhanced sympathetic activity), arterial calcification, vascular smooth muscle cell stiffness, and extracellular matrix glycosaminoglycans. Given the rapidly aging population of the United States, aortic stiffening will likely contribute to substantial CVD burden over the next 2-3 decades unless new therapeutic targets and interventions are identified to prevent the potential avalanche of clinical sequelae related to age-related aortic stiffness.
Collapse
Affiliation(s)
- Gary L Pierce
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa
| | - Thais A Coutinho
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Divisions of Cardiology and Cardiac Prevention and Rehabilitation, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Lyndsey E DuBose
- Division of Geriatrics, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony J Donato
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
- Department of Biochemistry, University of Utah, Salt Lake City, Utah
- Geriatric Research Education and Clinical Center, VA Salt Lake City, Salt Lake City, Utah
| |
Collapse
|
32
|
Custodero C, Ciavarella A, Panza F, Gnocchi D, Lenato GM, Lee J, Mazzocca A, Sabbà C, Solfrizzi V. Role of inflammatory markers in the diagnosis of vascular contributions to cognitive impairment and dementia: a systematic review and meta-analysis. GeroScience 2022; 44:1373-1392. [PMID: 35486344 PMCID: PMC9213626 DOI: 10.1007/s11357-022-00556-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/23/2022] [Indexed: 12/29/2022] Open
Abstract
Vascular contribution to cognitive impairment and dementia (VCID) is a clinical label encompassing a wide range of cognitive disorders progressing from mild to major vascular cognitive impairment (VCI), which is also defined as vascular dementia (VaD). VaD diagnosis is mainly based on clinical and imaging findings. Earlier biomarkers are needed to identify subjects at risk to develop mild VCI and VaD. In the present meta-analysis, we comprehensively evaluated the role of inflammatory biomarkers in differential diagnosis between VaD and Alzheimer’s disease (AD), and assessed their prognostic value on predicting VaD incidence. We collected literature until January 31, 2021, assessing three inflammatory markers [interleukin(IL)-6, C-reactive protein (CRP), tumor necrosis factor (TNF)-α] from blood or cerebrospinal fluid (CSF) samples. Thirteen cross-sectional and seven prospective studies were included. Blood IL-6 levels were cross-sectionally significantly higher in people with VaD compared to AD patients (SMD: 0.40, 95% CI: 0.18 to 0.62) with low heterogeneity (I2: 41%, p = 0.13). Higher IL-6 levels were also associated to higher risk of incident VaD (relative risk: 1.28, 95% CI: 1.03 to 1.59, I2: 0%). IL-6 in CSF was significantly higher in people with VaD compared to healthy subjects (SMD: 0.77, 95% CI: 0.17 to 1.37, I2: 70%), and not compared to AD patients, but due to limited evidence and high inconsistency across studies, we could not draw definite conclusion. Higher blood IL-6 levels might represent a useful biomarker able to differentiate people with VaD from those with AD and might be correlated with higher risk of future VaD.
Collapse
Affiliation(s)
- Carlo Custodero
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Alessandro Ciavarella
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy.,Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, A. Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Francesco Panza
- Population Health Unit-"Salus In Apulia Study", National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Davide Gnocchi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Gennaro M Lenato
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Juhan Lee
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Antonio Mazzocca
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Carlo Sabbà
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
33
|
Babcock MC, DuBose LE, Witten TL, Stauffer BL, Hildreth KL, Schwartz RS, Kohrt WM, Moreau KL. Oxidative Stress and Inflammation Are Associated With Age-Related Endothelial Dysfunction in Men With Low Testosterone. J Clin Endocrinol Metab 2022; 107:e500-e514. [PMID: 34597384 PMCID: PMC8764347 DOI: 10.1210/clinem/dgab715] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Vascular aging, including endothelial dysfunction secondary to oxidative stress and inflammation, increases the risk for age-associated cardiovascular disease (CVD). Low testosterone in middle-aged/older men is associated with increased CVD risk. OBJECTIVE We hypothesized that low testosterone contributes to age-associated endothelial dysfunction, related in part to greater oxidative stress and inflammation. METHODS This cross-sectional study included 58 healthy, nonsmoking men categorized as young (N = 20; age 29 ± 4 years; testosterone 500 ± 58 ng/dL), middle-aged/older with higher testosterone (N = 20; age 60 ± 6 years; testosterone 512 ± 115 ng/dL), and middle-aged/older lower testosterone (N = 18; age 59 ± 8 years; testosterone 269 ± 48 ng/dL). Brachial artery flow-mediated dilation (FMDBA) was measured during acute infusion of saline (control) and vitamin C (antioxidant). Markers of oxidative stress (total antioxidant status and oxidized low-density lipoprotein cholesterol), inflammation (interleukin [IL]-6 and C-reactive protein [CRP]), and androgen deficiency symptoms were also examined. RESULTS During saline, FMDBA was reduced in middle-aged/older compared with young, regardless of testosterone status (P < 0.001). FMDBA was reduced in middle-aged/older lower testosterone (3.7% ± 2.0%) compared with middle-aged/older higher testosterone (5.7% ± 2.2%; P = 0.021), independent of symptoms. Vitamin C increased FMDBA (to 5.3% ± 1.6%; P = 0.022) in middle-aged/older lower testosterone but had no effect in young (P = 0.992) or middle-aged/older higher testosterone (P = 0.250). FMDBA correlated with serum testosterone (r = 0.45; P < 0.001), IL-6 (r = -0.41; P = 0.002), and CRP (r = -0.28; P = 0.041). CONCLUSION Healthy middle-aged/older men with low testosterone appear to have greater age-associated endothelial dysfunction, related in part to greater oxidative stress and inflammation. These data suggest that low testosterone concentrations may contribute to accelerated vascular aging in men.
Collapse
Affiliation(s)
- Matthew C Babcock
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lyndsey E DuBose
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Teresa L Witten
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brian L Stauffer
- Division of Cardiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Division of Cardiology, Denver Health Medical Center, Denver, CO 80045, USA
| | - Kerry L Hildreth
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Robert S Schwartz
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Affairs Eastern Colorado Geriatric Research, Educational and Clinical Center, Denver, CO 80045, USA
| | - Wendy M Kohrt
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Affairs Eastern Colorado Geriatric Research, Educational and Clinical Center, Denver, CO 80045, USA
| | - Kerrie L Moreau
- Division of Geriatric Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Affairs Eastern Colorado Geriatric Research, Educational and Clinical Center, Denver, CO 80045, USA
- Correspondence: Kerrie L. Moreau, Ph.D., University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Geriatric Medicine, 12631 East 17th Ave., Mail Stop B179, Aurora, CO 80045, USA.
| |
Collapse
|
34
|
Wu J, Xu W, Wu W, Xu J, Zheng S, Shentu X, Chen X. Cataract-causing mutation R48C increases γA-crystallin susceptibility to oxidative stress and ultraviolet radiation. Int J Biol Macromol 2022; 194:688-694. [PMID: 34826455 DOI: 10.1016/j.ijbiomac.2021.11.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 01/16/2023]
Abstract
Among all congenital cataracts caused by genetic mutations, approximately half are caused by a mutation in crystallin genes, and accounts the leading cause of blindness in children globally. In this study, we investigated the underlying molecular mechanism of R48C mutation (c.142C > T; p.[Arg48Cys]) of γA-crystallin in a Mexican-Mestizo descent family causing congenital cataracts. We purified γA-crystallin wild-type (WT) and R48C mutant and compared their structural characteristics and biophysical properties by Spectroscopic experiments and environmental stress (oxidative stress, ultraviolet irradiation, pH disorders, thermal shock, or chemical denaturation). The R48C mutant did not affect the secondary and tertiary structure of monomer γA-crystallin, nor did it affect its stability to heat shock and chemicals. However, the R48C mutant destroys the oxidative stability of γA-crystallin, which makes the protein more prone to aggregation and precipitation under oxidative conditions. These might be the pathogenesis of γA-crystallin R48C mutant related to congenital cataract and help to develop anti-cataract strategies from the perspective of γA-crystallin.
Collapse
Affiliation(s)
- Jing Wu
- Department of Ophthalmology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Lab of Ophthalmology, 88 Jiefang Road, Hangzhou, Zhejiang, China; Department of Ophthalmology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang, China
| | - Wanyue Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kaixuan Road, Hangzhou, Zhejiang, China
| | - Wei Wu
- Department of Ophthalmology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Lab of Ophthalmology, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Jingjie Xu
- Department of Ophthalmology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Lab of Ophthalmology, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Sifan Zheng
- GKT School of Medical Education, King's College London, London SE1 1UL, United Kingdom
| | - Xingchao Shentu
- Department of Ophthalmology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Lab of Ophthalmology, 88 Jiefang Road, Hangzhou, Zhejiang, China.
| | - Xiangjun Chen
- Department of Ophthalmology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Lab of Ophthalmology, 88 Jiefang Road, Hangzhou, Zhejiang, China; Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kaixuan Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
35
|
Labissiere X, Zigmond ZM, Challa A, Montoya C, Manzur-Pineda K, Abraham A, Tabbara M, Salama A, Pan Y, Salman LH, Yang X, Vazquez-Padron RI, Martinez L. Vein morphometry in end-stage kidney disease: Teasing out the contribution of age, comorbidities, and vintage to chronic wall remodeling. Front Cardiovasc Med 2022; 9:1005030. [PMID: 36419492 PMCID: PMC9676677 DOI: 10.3389/fcvm.2022.1005030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Background Chronic kidney disease (CKD) is a highly comorbid condition with significant effects on vascular health and remodeling. Upper extremity veins are important in end-stage kidney disease (ESKD) due to their potential use to create vascular accesses. However, unlike arteries, the contribution of CKD-associated factors to the chronic remodeling of veins has been barely studied. Methods We measured morphometric parameters in 315 upper extremity veins, 131 (85% basilic) from stage 5 CKD/ESKD patients and 184 (89% basilic) from non-CKD organ donors. Associations of demographic and clinical characteristics with intimal hyperplasia (IH) and medial fibrosis were evaluated using multivariate regression models. Results The study cohort included 33% females, 30% blacks, 32% Hispanics, and 37% whites. Over 60% had hypertension, and 25% had diabetes independent of CKD status. Among kidney disease participants, 26% had stage 5 CKD, while 22 and 52% had ESKD with and without history of a previous arteriovenous fistula/graft (AVF/AVG), respectively. Intimal hyperplasia was associated with older age (β = 0.13 per year, confidence interval [CI] = 0.002-0.26), dialysis vintage > 12 months (β = 0.22, CI = 0.09-0.35), and previous AVF/AVG creation (β = 0.19, CI = 0.06-0.32). Upper quartile values of IH were significantly associated with diabetes (odds ratio [OR] = 2.02, CI = 1.08-3.80), which demonstrated an additive effect with previous AVF/AVG history and longer vintage in exacerbating IH. Medial fibrosis also increased as a function of age (β = 0.17, CI = 0.04-0.30) and among patients with diabetes (β = 0.15, CI = 0.03-0.28). Age was the predominant factor predicting upper quartile values of fibrosis (OR = 1.03 per year, CI = 1.01-1.05) independent of other comorbidities. Conclusion Age and diabetes are the most important risk factors for chronic development of venous IH and fibrosis independent of CKD status. Among kidney disease patients, longer dialysis vintage, and history of a previous AVF/AVG are strong predictors of IH.
Collapse
Affiliation(s)
- Xochilt Labissiere
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Zachary M Zigmond
- Bruce W. Carter Veterans Affairs Medical Center, Miami, FL, United States
| | - Akshara Challa
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Christopher Montoya
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Karen Manzur-Pineda
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Amalia Abraham
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Marwan Tabbara
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Alghidak Salama
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Yue Pan
- Department of Public Health Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Loay H Salman
- Division of Nephrology, Albany Medical College, Albany, NY, United States
| | - Xiaofeng Yang
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States.,Bruce W. Carter Veterans Affairs Medical Center, Miami, FL, United States
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
36
|
Molnár AÁ, Nádasy GL, Dörnyei G, Patai BB, Delfavero J, Fülöp GÁ, Kirkpatrick AC, Ungvári Z, Merkely B. The aging venous system: from varicosities to vascular cognitive impairment. GeroScience 2021; 43:2761-2784. [PMID: 34762274 PMCID: PMC8602591 DOI: 10.1007/s11357-021-00475-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/12/2021] [Indexed: 10/25/2022] Open
Abstract
Aging-induced pathological alterations of the circulatory system play a critical role in morbidity and mortality of older adults. While the importance of cellular and molecular mechanisms of arterial aging for increased cardiovascular risk in older adults is increasingly appreciated, aging processes of veins are much less studied and understood than those of arteries. In this review, age-related cellular and morphological alterations in the venous system are presented. Similarities and dissimilarities between arterial and venous aging are highlighted, and shared molecular mechanisms of arterial and venous aging are considered. The pathogenesis of venous diseases affecting older adults, including varicose veins, chronic venous insufficiency, and deep vein thrombosis, is discussed, and the potential contribution of venous pathologies to the onset of vascular cognitive impairment and neurodegenerative diseases is emphasized. It is our hope that a greater appreciation of the cellular and molecular processes of vascular aging will stimulate further investigation into strategies aimed at preventing or retarding age-related venous pathologies.
Collapse
Affiliation(s)
- Andrea Ágnes Molnár
- Heart and Vascular Center, Semmelweis University, Városmajor Street 68, 1121, Budapest, Hungary.
| | | | - Gabriella Dörnyei
- Department of Morphology and Physiology, Health Sciences Faculty, Semmelweis University, Budapest, Hungary
| | | | - Jordan Delfavero
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center On Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gábor Áron Fülöp
- Heart and Vascular Center, Semmelweis University, Városmajor Street 68, 1121, Budapest, Hungary
| | - Angelia C Kirkpatrick
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Veterans Affairs Medical Center, 921 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Zoltán Ungvári
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center On Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Városmajor Street 68, 1121, Budapest, Hungary
| |
Collapse
|
37
|
Greiten LE, Zhang B, Roos CM, Hagler M, Jahns FP, Miller JD. Sirtuin 6 Protects Against Oxidative Stress and Vascular Dysfunction in Mice. Front Physiol 2021; 12:753501. [PMID: 34744793 PMCID: PMC8564013 DOI: 10.3389/fphys.2021.753501] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/28/2021] [Indexed: 01/05/2023] Open
Abstract
Objective: Sirtuin deacetylases are major regulators of organismal aging, and while depletion of sirtuin 6 (SIRT6) in mice results in a profound progeroid phenotype, the role of SIRT6 in the regulation of vasomotor function is unknown. Thus, our objective was to test the hypothesis that reductions in SIRT6 elicit endothelial dysfunction in young, genetically altered mice. Results and Approach: We used young (3 month old), littermate-matched, SIRT6 wild-type (WT), and SIRT6 heterozygous (HET) mice. SIRT6 expression (qRT-PCR) was reduced by 50% in HET mice. Carotid vessel responses to acetylcholine, sodium nitroprusside, U46619, and serotonin were examined in isolated organ chamber baths. Relaxation in response to acetylcholine (ACH) was impaired in HET mice compared to littermate-matched WT controls (67 ± 3% versus 76 ± 3%, respectively; p < 0.05), while responses to sodium nitroprusside were unchanged. Short-term incubation of carotid rings with the NAD(P)H oxidase inhibitor, apocynin, significantly improved in vessels from HET mice but not their WT littermates. Peak tension generated in response to either U46619 or serotonin was significantly blunted in HET mice compared to their WT littermates. Conclusion: These data suggest that SIRT6 is a key regulator of vasomotor function in conduit vessels. More specifically, we propose that SIRT6 serves as a tonic suppressor of NAD(P)H oxidase expression and activation, as inhibition of NAD(P)H oxidase improved endothelial function in SIRT6 haploinsufficient mice. Collectively, SIRT6 activation and/or histone acetyltransferase inhibition may be useful therapeutic approaches to reduce endothelial dysfunction and combat age-associated cardiovascular disease.
Collapse
Affiliation(s)
| | - Bin Zhang
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Carolyn M Roos
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Michael Hagler
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Jordan D Miller
- Department of Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Biomedical Engineering and Physiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
38
|
Francisco MA, Lee JF, Barrett-O'Keefe Z, Groot HJ, Ratchford SM, Bunsawat K, Alpenglow JK, Ryan JJ, Nativi JN, Richardson RS, Wray DW. Locomotor Muscle Microvascular Dysfunction in Heart Failure With Preserved Ejection Fraction. Hypertension 2021; 78:1750-1759. [PMID: 34719934 DOI: 10.1161/hypertensionaha.121.17875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Michael A Francisco
- Department of Internal Medicine (M.A.F., J.F.L., K.B., J.J.R., J.N.N., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.)
| | - Joshua F Lee
- Department of Internal Medicine (M.A.F., J.F.L., K.B., J.J.R., J.N.N., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.)
| | - Zachary Barrett-O'Keefe
- Department of Nutrition and Integrative Physiology (Z.B.-O., H.J.G., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.)
| | - H Jonathan Groot
- Department of Nutrition and Integrative Physiology (Z.B.-O., H.J.G., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.)
| | - Stephen M Ratchford
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.).,Department of Health and Exercise Science, Appalachian State University, Boone, NC (S.M.R.)
| | - Kanokwan Bunsawat
- Department of Internal Medicine (M.A.F., J.F.L., K.B., J.J.R., J.N.N., R.S.R., D.W.W.), University of Utah, Salt Lake City
| | - Jeremy K Alpenglow
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.)
| | - John J Ryan
- Department of Internal Medicine (M.A.F., J.F.L., K.B., J.J.R., J.N.N., R.S.R., D.W.W.), University of Utah, Salt Lake City
| | - Jose N Nativi
- Department of Internal Medicine (M.A.F., J.F.L., K.B., J.J.R., J.N.N., R.S.R., D.W.W.), University of Utah, Salt Lake City
| | - Russell S Richardson
- Department of Internal Medicine (M.A.F., J.F.L., K.B., J.J.R., J.N.N., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Department of Nutrition and Integrative Physiology (Z.B.-O., H.J.G., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.)
| | - D Walter Wray
- Department of Internal Medicine (M.A.F., J.F.L., K.B., J.J.R., J.N.N., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Department of Nutrition and Integrative Physiology (Z.B.-O., H.J.G., R.S.R., D.W.W.), University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, VA Medical Center, Salt Lake City, UT (M.A.F., J.F.L., Z.B.-O., H.J.G., S.M.R., J.K.A., R.S.R., D.W.W.)
| |
Collapse
|
39
|
Lefferts EC, Hibner BA, Lefferts WK, Lima NS, Baynard T, Haus JM, Lane‐Cordova AD, Phillips SA, Fernhall B. Oral vitamin C restores endothelial function during acute inflammation in young and older adults. Physiol Rep 2021; 9:e15104. [PMID: 34762777 PMCID: PMC8582295 DOI: 10.14814/phy2.15104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 11/24/2022] Open
Abstract
Oxidative stress has been linked to reductions in vascular function during acute inflammation in young adults; however, the effect of acute inflammation on vascular function with aging is inconclusive. The aim of this study was to determine if oral antioxidant administration eliminates vascular dysfunction during acute inflammation in young and older adults. Brachial flow-mediated dilation (FMD) and carotid-femoral pulse wave velocity (PWV) were measured in nine young (3 male, 24 ± 4 yrs, 26.2 ± 4.9 kg/m2 ) and 16 older (13 male, 64 ± 5 yrs, 25.8 ± 3.2 kg/m2 ) adults before and 2-h after oral consumption of 2 g of vitamin C. The vitamin C protocol was completed at rest and 24 h after acute inflammation was induced via the typhoid vaccine. Venous blood samples were taken to measure markers of inflammation and vitamin C. Both interleukin-6 (Δ+0.7 ± 1.8 pg/ml) and C-reactive protein (Δ+1.9 ± 3.1 mg/L) were increased at 24 h following the vaccine (p < 0.01). There was no change in FMD or PWV following vitamin C administration at rest (p > 0.05). FMD was lower in all groups during acute inflammation (Δ-1.4 ± 1.9%, p < 0.01), with no changes in PWV (Δ-0.0 ± 0.9 m/s, p > 0.05). Vitamin C restored FMD back to initial values in young and older adults during acute inflammation (Δ+1.0 ± 1.8%, p < 0.01) with no change in inflammatory markers or PWV (p > 0.05). In conclusion, oral vitamin C restored endothelial function during acute inflammation in young and older adults, with no effect on aortic stiffness. The effect of vitamin C on endothelial function did not appear to be due to reductions in inflammatory markers. The exact mechanisms should be further investigated.
Collapse
Affiliation(s)
- Elizabeth C. Lefferts
- Department of Kinesiology and NutritionUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of KinesiologyIowa State UniversityAmesIowaUSA
| | - Brooks A. Hibner
- Department of Kinesiology and NutritionUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Wesley K. Lefferts
- Department of Kinesiology and NutritionUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of KinesiologyIowa State UniversityAmesIowaUSA
| | - Natalia S. Lima
- Department of Kinesiology and NutritionUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Tracy Baynard
- Department of Kinesiology and NutritionUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Jacob M. Haus
- School of KinesiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Abbi D. Lane‐Cordova
- Department of Exercise ScienceArnold School of Public HealthUniversity of South CarolinaColumbiaSouth CarolinaUSA
| | - Shane A. Phillips
- Department of Physical TherapyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Bo Fernhall
- Department of Kinesiology and NutritionUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
40
|
Dynamic Crosstalk between Vascular Smooth Muscle Cells and the Aged Extracellular Matrix. Int J Mol Sci 2021; 22:ijms221810175. [PMID: 34576337 PMCID: PMC8468233 DOI: 10.3390/ijms221810175] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/15/2023] Open
Abstract
Vascular aging is accompanied by the fragmentation of elastic fibers and collagen deposition, leading to reduced distensibility and increased vascular stiffness. A rigid artery facilitates elastin to degradation by MMPs, exposing vascular cells to greater mechanical stress and triggering signaling mechanisms that only exacerbate aging, creating a self-sustaining inflammatory environment that also promotes vascular calcification. In this review, we highlight the role of crosstalk between smooth muscle cells and the vascular extracellular matrix (ECM) and how aging promotes smooth muscle cell phenotypes that ultimately lead to mechanical impairment of aging arteries. Understanding the underlying mechanisms and the role of associated changes in ECM during aging may contribute to new approaches to prevent or delay arterial aging and the onset of cardiovascular diseases.
Collapse
|
41
|
Time-efficient, high-resistance inspiratory muscle strength training for cardiovascular aging. Exp Gerontol 2021; 154:111515. [PMID: 34389471 DOI: 10.1016/j.exger.2021.111515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/29/2021] [Accepted: 08/09/2021] [Indexed: 01/14/2023]
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death in developed and developing societies and aging is the primary risk factor for CVD. Much of the increased risk of CVD in midlife/older adults (i.e., adults aged 50 years and older) is due to increases in blood pressure, vascular endothelial dysfunction and stiffening of the large elastic arteries. Aerobic exercise training is an effective lifestyle intervention to improve CV function and decrease CVD risk with aging. However, <40% of midlife/older adults meet guidelines for aerobic exercise, due to time availability-related barriers and other obstacles to adherence. Therefore, there is a need for new lifestyle interventions that not only improve CV function with aging but also promote adherence. High-resistance inspiratory muscle strength training (IMST) is an emerging, time-efficient (5 min/day) lifestyle intervention. Early research suggests high-resistance IMST may promote adherence, lower blood pressure and potentially improve vascular endothelial function. However, additional investigation will be required to more definitively establish high-resistance IMST as a healthy lifestyle intervention for CV aging. This review will summarize the current evidence on high-resistance IMST for improving CV function with aging and will identify key research gaps and future directions.
Collapse
|
42
|
Nicosia RF, Ligresti G, Caporarello N, Akilesh S, Ribatti D. COVID-19 Vasculopathy: Mounting Evidence for an Indirect Mechanism of Endothelial Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1374-1384. [PMID: 34033751 PMCID: PMC8141344 DOI: 10.1016/j.ajpath.2021.05.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
Patients with coronavirus disease 2019 (COVID-19) who are critically ill develop vascular complications characterized by thrombosis of small, medium, and large vessels. Dysfunction of the vascular endothelium due to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been implicated in the pathogenesis of the COVID-19 vasculopathy. Although initial reports suggested that endothelial injury was caused directly by the virus, recent studies indicate that endothelial cells do not express angiotensin-converting enzyme 2, the receptor that SARS-CoV-2 uses to gain entry into cells, or express it at low levels and are resistant to the infection. These new findings, together with the observation that COVID-19 triggers a cytokine storm capable of injuring the endothelium and disrupting its antithrombogenic properties, favor an indirect mechanism of endothelial injury mediated locally by an augmented inflammatory reaction to infected nonendothelial cells, such as the bronchial and alveolar epithelium, and systemically by the excessive immune response to infection. Herein we review the vascular pathology of COVID-19 and critically discuss the potential mechanisms of endothelial injury in this disease.
Collapse
Affiliation(s)
- Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington.
| | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neuroscienze e Organi di Senso (SMBNOS), Universita' degli Studi Aldo Moro, Policlinico, Bari, Italy
| |
Collapse
|
43
|
Trott DW, Machin DR, Phuong TTT, Adeyemo AO, Bloom SI, Bramwell RC, Sorensen ES, Lesniewski LA, Donato AJ. T cells mediate cell non-autonomous arterial ageing in mice. J Physiol 2021; 599:3973-3991. [PMID: 34164826 PMCID: PMC8425389 DOI: 10.1113/jp281698] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/21/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Increased large artery stiffness and impaired endothelium-dependent dilatation occur with advanced age. We sought to determine whether T cells mechanistically contribute to age-related arterial dysfunction. We found that old mice exhibited greater proinflammatory T cell accumulation around both the aorta and mesenteric arteries. Pharmacologic depletion or genetic deletion of T cells in old mice resulted in ameliorated large artery stiffness and greater endothelium-dependent dilatation compared with mice with T cells intact. ABSTRACT Ageing of the arteries is characterized by increased large artery stiffness and impaired endothelium-dependent dilatation. T cells contribute to hypertension in acute rodent models but whether they contribute to chronic age-related arterial dysfunction is unknown. To determine whether T cells directly mediate age-related arterial dysfunction, we examined large elastic artery and resistance artery function in young (4-6 months) and old (22-24 months) wild-type mice treated with anti-CD3 F(ab'2) fragments to deplete T cells (150 μg, i.p. every 7 days for 28 days) or isotype control fragments. Old mice exhibited greater numbers of T cells in both aorta and mesenteric vasculature when compared with young mice. Old mice treated with anti-CD3 fragments exhibited depletion of T cells in blood, spleen, aorta and mesenteric vasculature. Old mice also exhibited greater numbers of aortic and mesenteric IFN-γ and TNF-α-producing T cells when compared with young mice. Old control mice exhibited greater large artery stiffness and impaired resistance artery endothelium-dependent dilatation in comparison with young mice. In old mice, large artery stiffness was ameliorated with anti-CD3 treatment. Anti-CD3-treated old mice also exhibited greater endothelium-dependent dilatation than age-matched controls. We also examined arterial function in young and old Rag-1-/- mice, which lack lymphocytes. Rag-1-/- mice exhibited blunted increases in large artery stiffness with age compared with wild-type mice. Old Rag-1-/- mice also exhibited greater endothelium-dependent dilatation compared with old wild-type mice. Collectively, these results demonstrate that T cells play an important role in age-related arterial dysfunction.
Collapse
Affiliation(s)
- Daniel W Trott
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Kinesiology, University of Texas at Arlington, Texas, USA
| | - Daniel R Machin
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Tam T T Phuong
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - AdeLola O Adeyemo
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Samuel I Bloom
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - R Colton Bramwell
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Eric S Sorensen
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Lisa A Lesniewski
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
- Geriatrics Research Education and Clinical Center, Veteran's Affairs Medical Center, Salt Lake City, Utah, USA
| | - Anthony J Donato
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
- Geriatrics Research Education and Clinical Center, Veteran's Affairs Medical Center, Salt Lake City, Utah, USA
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
44
|
Govindasamy V, Rajendran A, Lee ZX, Ooi GC, Then KY, Then KL, Gayathri M, Kumar Das A, Cheong SK. The potential role of mesenchymal stem cells in modulating antiageing process. Cell Biol Int 2021; 45:1999-2016. [PMID: 34245637 DOI: 10.1002/cbin.11652] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/24/2021] [Accepted: 06/17/2021] [Indexed: 12/19/2022]
Abstract
Ageing and age-related diseases share some basic origin that largely converges on inflammation. Precisely, it boils down to a common pathway characterised by the appearance of a fair amount of proinflammatory cytokines known as inflammageing. Among the proposed treatment for antiageing, MSCs gained attention in recent years. Since mesenchymal stem cells (MSCs) can differentiate itself into a myriad of terminal cells, previously it was believed that these cells migrate to the site of injury and perform their therapeutic effect. However, with the more recent discovery of huge amounts of paracrine factors secreted by MSCs, it is now widely accepted that these cells do not engraft upon transplantation but rather unveil their benefits through excretion of bioactive molecules namely those involved in inflammatory and immunomodulatory activities. Conversely, the true function of these paracrine changes has not been thoroughly investigated all these years. Hence, this review will describe in detail on ways MSCs may capitalize its paracrine properties in modulating antiageing process. Through a comprehensive literature search various elements in the antiageing process, we aim to provide a novel treatment perspective of MSCs in antiageing related clinical conditions.
Collapse
Affiliation(s)
- Vijayendran Govindasamy
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Abilashini Rajendran
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Zhi-Xin Lee
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Ghee-Chien Ooi
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Kong-Yong Then
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia.,Brighton Healthcare (Bio-X Healthcare Sdn Bhd), Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Khong-Lek Then
- Research and Development Department, CryoCord Sdn Bhd, Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Merilynn Gayathri
- Brighton Healthcare (Bio-X Healthcare Sdn Bhd), Bio-X Centre, Cyberjaya, Selangor, Malaysia
| | - Anjan Kumar Das
- Deparment of Surgery, IQ City Medical College, Durgapur, West Bengal, India
| | - Soon-Keng Cheong
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman (UTAR), Kajang, Selangor, Malaysia
| |
Collapse
|
45
|
Chala N, Moimas S, Giampietro C, Zhang X, Zambelli T, Exarchos V, Nazari-Shafti TZ, Poulikakos D, Ferrari A. Mechanical Fingerprint of Senescence in Endothelial Cells. NANO LETTERS 2021; 21:4911-4920. [PMID: 34081865 DOI: 10.1021/acs.nanolett.1c00064] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Endothelial senescence entails alterations of the healthy cell phenotype, which accumulate over time and contribute to cardiovascular disease. Mechanical aspects regulating cell adhesion, force generation, and the response to flow contribute to the senescence-associated drift; however, they remain largely unexplored. Here, we exploit force microscopy to resolve variations of the cell anchoring to the substrate and the tractions generated upon aging in the nanonewton (nN) range. Senescent endothelial cells display a multifold increase in the levels of basal adhesion and force generation supported by mature and strong focal adhesions. The enhanced mechanical interaction with the substrate yields static endothelial monolayers that polarize in response to flow but fail the process of coordinated cell shape remodeling and reorientation. The emerging picture indicates that senescence reinforces the local cell interaction with the substrate and may therefore prevent endothelial denudation; however, it compromises the ability to functionally adapt to the local hemodynamic conditions.
Collapse
Affiliation(s)
- Nafsika Chala
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zürich, Switzerland
| | - Silvia Moimas
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zürich, Switzerland
| | - Costanza Giampietro
- Experimental Continuum Mechanics, EMPA, Swiss Federal Laboratories for Material Science and Technologies, Überlandstrasse 129, 8600 Dübendorf, Switzerland
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Leonhardstrasse 21, 8092 Zürich, Switzerland
| | - Xinyu Zhang
- Laboratory of Biosensors and Bioelectronics, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 35, 8092Zürich, Switzerland
| | - Tomaso Zambelli
- Laboratory of Biosensors and Bioelectronics, Department of Information Technology and Electrical Engineering, ETH Zurich, Gloriastrasse 35, 8092Zürich, Switzerland
| | - Vasileios Exarchos
- German Heart Center Berlin, Department for Cardiovascular and Thoracic Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Timo Z Nazari-Shafti
- German Heart Center Berlin, Department for Cardiovascular and Thoracic Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center for Regenerative Therapies, Föhrer Strasse 15, 13353 Berlin, Germany
| | - Dimos Poulikakos
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zürich, Switzerland
| | - Aldo Ferrari
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, 8092 Zürich, Switzerland
- Experimental Continuum Mechanics, EMPA, Swiss Federal Laboratories for Material Science and Technologies, Überlandstrasse 129, 8600 Dübendorf, Switzerland
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Leonhardstrasse 21, 8092 Zürich, Switzerland
| |
Collapse
|
46
|
Han A, Kim JY, Kwak-Kim J, Lee SK. Menopause is an inflection point of age-related immune changes in women. J Reprod Immunol 2021; 146:103346. [PMID: 34147765 DOI: 10.1016/j.jri.2021.103346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
Elevated proinflammatory cytokines in postmenopausal women is considered as one of the causes increasing the incidence of chronic inflammatory diseases. However, the details of postmenopausal immune changes have not yet been fully revealed. Thus, we investigated age-related immune changes in women and compared immune responses in postmenopausal and reproductive-age women. A total of 34 postmenopausal women and 91 reproductive-age women were included in the study. After isolating peripheral blood mononuclear cells, analysis of immunophenotypes and intracellular cytokine profiles were done. The proportion of natural killer (NK) cells was significantly higher, and the ratio of TNF-α- to IL-10-producing CD3+CD4 + T cells (Th1 to Th2) and the ratio of Th17 cells to CD4+CD25+Foxp3+ regulatory T (Treg) cells (Th17 to Treg) were higher, in postmenopausal women than in reproductive-age women. The Treg cell proportion was negatively correlated with the Th1 and Th2 cell proportions in reproductive-age women but not in postmenopausal women. As age increased, the proportion of Tregs was increased in reproductive-age women (r = 0.302, p = 0.004), whereas the proportion of Th1 cells was increased in postmenopausal women (r = 0.466, p = 0.005). FSH levels showed a positive correlation with Fopx3+ T cell and Treg cell (p = 0.04, 0.053, respectively), whereas Th17/Treg ratio and Th1 cell showed negative correlation with FSH.(p = 0.045, 0.024, respectively). In conclusion, postmenopausal women have higher proinflammatory immune statuses, as demonstrated by increased proportions of NK, Th1, and Th17 cells, altered correlations among NK and T cell subsets, and compromised balances between effector T cell subsets.
Collapse
Affiliation(s)
- Aera Han
- Konyang University Myunggok Research Institute, Republic of Korea
| | - Jee Yun Kim
- Department of Obstetrics and Gynecology, Konyang University College of Medicine, Daejeon, Republic of Korea
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School at Rosalind Franklin University of Medicine and Science, Vernon Hills, USA; Department of Microbiology and Immunology, Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sung Ki Lee
- Konyang University Myunggok Research Institute, Republic of Korea; Department of Obstetrics and Gynecology, Konyang University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW To summarize the state of chronic, treated HIV infection and its contribution to accelerated aging, and to evaluate recent research relevant to the study and treatment of aging and senescence. RECENT FINDINGS Chronic treated HIV-1 infection is associated with significant risk of end-organ impairment, non-AIDS-associated malignancies, and accelerated physiologic aging. Coupled with the chronologic aging of the HIV-1-positive population, the development of therapies that target these processes is of great clinical importance. Age-related diseases are partly the result of cellular senescence. Both immune and nonimmune cell subsets are thought to mediate this senescent phenotype, a state of stable cell cycle arrest characterized by sustained release of pro-inflammatory mediators. Recent research in the field of aging has identified a number of 'senotherapeutics' to combat aging-related diseases, pharmacologic agents that act either by selectively promoting the death of senescent cells ('senolytics') or modifying senescent phenotype ('senomorphics'). SUMMARY Senescence is a hallmark of aging-related diseases that is characterized by stable cell cycle arrest and chronic inflammation. Chronic HIV-1 infection predisposes patients to aging-related illnesses and is similarly marked by a senescence-like phenotype. A better understanding of the role of HIV-1 in aging will inform the development of therapeutics aimed at eliminating senescent cells that drive accelerated physiologic aging.
Collapse
|
48
|
Tropea T, Mandalà M. Caloric restriction enhances vascular tone of cerebral and mesenteric resistance arteries in aged rats. Mech Ageing Dev 2021; 197:111520. [PMID: 34129890 DOI: 10.1016/j.mad.2021.111520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 11/27/2022]
Abstract
Vascular changes of tone and biomechanical properties induced by ageing increase the risk for cardiovascular diseases. Caloric restriction (CR) has been shown to protect against cardiovascular diseases and improve endothelial dysfunction in cerebral resistance arteries. We hypothesise that CR will enhance vascular tone and structural properties of cerebral resistance arteries and exert comparable beneficial effects on the systemic vasculature of aged rat model. Eighteen-month-old male Sprague-Dawley rats were feed either ad libitum or restricted to 60 % of calorie consumption up to 24 months of age, when body weight (BW) measurements were taken and functional and structural properties of resistance arteries were assessed using a pressure myograph. In cerebral arteries, CR increased myogenic tone (p < 0.001) and distensibility (p < 0.01) in response to intraluminal pressure and concentration-dependent constriction to KCl (p < 0.001). In mesenteric arteries constriction in response to KCl was increased (p < 0.0001) and wall thickness reduced (p < 0.01) in CR rats. BW was reduced (p < 0.0001) in FR rats. Our findings demonstrate that CR improves vascular tone of resistance arteries regardless the type of stimulus and independently of the vascular bed. CR may be a beneficial dietary approach to prevent age-related vascular diseases.
Collapse
Affiliation(s)
- Teresa Tropea
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, United Kingdom; Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy
| | - Maurizio Mandalà
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy; Department of Obstetrics, Gynecology and Reproductive Science, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
49
|
Costa J, Martins S, Ferreira PA, Cardoso AMS, Guedes JR, Peça J, Cardoso AL. The old guard: Age-related changes in microglia and their consequences. Mech Ageing Dev 2021; 197:111512. [PMID: 34022277 DOI: 10.1016/j.mad.2021.111512] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022]
Abstract
Among all major organs, the brain is one of the most susceptible to the inexorable effects of aging. Throughout the last decades, several studies in human cohorts and animal models have revealed a plethora of age-related changes in the brain, including reduced neurogenesis, oxidative damage, mitochondrial dysfunction and cell senescence. As the main immune effectors and first responders of the nervous tissue, microglia are at the center of these events. These cells experience irrevocable changes as a result from cumulative exposure to environmental triggers, such as stress, infection and metabolic dysregulation. The age-related immunosenescent phenotype acquired by microglia is characterized by profound modifications in their transcriptomic profile, secretome, morphology and phagocytic activity, which compromise both their housekeeping and defensive functions. As a result, aged microglia are no longer capable of establishing effective immune responses and sustaining normal synaptic activity, directly contributing to age-associated cognitive decline and neurodegeneration. This review discusses how lifestyle and environmental factors drive microglia dysfunction at the molecular and functional level, also highlighting possible interventions to reverse aging-associated damage to the nervous and immune systems.
Collapse
Affiliation(s)
- Jéssica Costa
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Solange Martins
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Pedro A Ferreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; PhD Program in Biosciences, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana M S Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Joana R Guedes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - João Peça
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana L Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
50
|
Inflammageing in the cardiovascular system: mechanisms, emerging targets, and novel therapeutic strategies. Clin Sci (Lond) 2021; 134:2243-2262. [PMID: 32880386 DOI: 10.1042/cs20191213] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022]
Abstract
In the elderly population, pathological inflammation has been associated with ageing-associated diseases. The term 'inflammageing', which was used for the first time by Franceschi and co-workers in 2000, is associated with the chronic, low-grade, subclinical inflammatory processes coupled to biological ageing. The source of these inflammatory processes is debated. The senescence-associated secretory phenotype (SASP) has been proposed as the main origin of inflammageing. The SASP is characterised by the release of inflammatory cytokines, elevated activation of the NLRP3 inflammasome, altered regulation of acetylcholine (ACh) nicotinic receptors, and abnormal NAD+ metabolism. Therefore, SASP may be 'druggable' by small molecule therapeutics targeting those emerging molecular targets. It has been shown that inflammageing is a hallmark of various cardiovascular diseases, including atherosclerosis, hypertension, and adverse cardiac remodelling. Therefore, the pathomechanism involving SASP activation via the NLRP3 inflammasome; modulation of NLRP3 via α7 nicotinic ACh receptors; and modulation by senolytics targeting other proteins have gained a lot of interest within cardiovascular research and drug development communities. In this review, which offers a unique view from both clinical and preclinical target-based drug discovery perspectives, we have focused on cardiovascular inflammageing and its molecular mechanisms. We have outlined the mechanistic links between inflammageing, SASP, interleukin (IL)-1β, NLRP3 inflammasome, nicotinic ACh receptors, and molecular targets of senolytic drugs in the context of cardiovascular diseases. We have addressed the 'druggability' of NLRP3 and nicotinic α7 receptors by small molecules, as these proteins represent novel and exciting targets for therapeutic interventions targeting inflammageing in the cardiovascular system and beyond.
Collapse
|